US20010027197A1 - Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family - Google Patents

Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family Download PDF

Info

Publication number
US20010027197A1
US20010027197A1 US09/824,606 US82460601A US2001027197A1 US 20010027197 A1 US20010027197 A1 US 20010027197A1 US 82460601 A US82460601 A US 82460601A US 2001027197 A1 US2001027197 A1 US 2001027197A1
Authority
US
United States
Prior art keywords
carbon atoms
amino
carbon
pyrimidine
mono
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US09/824,606
Other versions
US6455534B2 (en
Inventor
Alexander Bridges
William Denny
David Fry
Alan Kraker
Robert Meyer
Gordon Rewcastle
Andrew Thompson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/358,351 external-priority patent/US5654307A/en
Application filed by Individual filed Critical Individual
Priority to US09/824,606 priority Critical patent/US6455534B2/en
Publication of US20010027197A1 publication Critical patent/US20010027197A1/en
Application granted granted Critical
Publication of US6455534B2 publication Critical patent/US6455534B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/16Masculine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the present invention relates to bicyclic heteroaromatic compounds which inhibit the epidermal growth factor receptor and related receptors and, in particular, their tyrosine kinase enzymic activity.
  • Cancer is generally a disease of the intracellular signalling system, or signal transduction mechanism.
  • Cells receive instructions from many extracellular sources, instructing them to either proliferate or not to proliferate.
  • the purpose of the signal transduction system is to receive these and other signals at the cell surface, get them into the cell, and then pass the signals on to the nucleus, the cytoskeleton, and transport and protein synthesis machinery.
  • the most common cause of cancer is a series of defects, either in these proteins, when they are mutated, or in the regulation of the quantity of the protein in the cell such that it is over or under produced.
  • Most often, there are key lesions in the cell which lead to a constitutive state whereby the cell nucleus receives a signal to proliferate, when this signal is not actually present.
  • the cell may start to produce an authentic growth factor for its own receptors when it should not, the so-called autocrine loop mechanism. Mutations to the cell surface receptors, which usually signal into the cell by means of tyrosine kinases, can lead to activation of the kinase in the absence of ligand, and passing of a signal which is not really there. Alternatively, many surface kinases can be overexpressed on the cell surface leading to an inappropriately strong response to a weak signal. There are many levels inside the cell at which mutation or overexpression can lead to the same spurious signal arising in the cell, and there are many other kinds of signalling defect involved in cancer.
  • This invention touches upon cancers which are driven by the three mechanisms just described, and which involve cell surface receptors of the epidermal growth factor receptor tyrosine kinase family (EGFR).
  • EGFR epidermal growth factor receptor tyrosine kinase family
  • This family consists of the EGF receptor (also known as Erb-B1), the Erb-B2 receptor, and its constituitively active oncoprotein mutant Neu, the Erb-B3 receptor and the Erb-B4 receptor.
  • EGF receptor also known as Erb-B1
  • Erb-B2 receptor also known as Erb-B2 receptor
  • its constituitively active oncoprotein mutant Neu the Erb-B3 receptor
  • Erb-B4 receptor the Erb-B4 receptor
  • the EGFR has as its two most important ligands Epidermal Growth Factor (EGF) and Transforming Growth Factor alpha (TGFalpha).
  • EGF Epidermal Growth Factor
  • TGFalpha Transforming Growth Factor alpha
  • the receptors appear to have only minor functions in adult humans, but are apparently implicated in the disease process of a large portion of all cancers, especially colon and breast cancer.
  • the closely related Erb-B2 Erb-B3 and Erb-B4 receptors have a family of Heregulins as their major ligands, and receptor overexpression and mutation have been unequivocally demonstrated as the major risk factor in poor prognosis breast cancer.
  • the proliferative skin disease psoriasis has no good cure at present. It is often treated by anticancer agents such as methotrexate, which have very serious side effects, and which are not very effective at the toxicity-limited doses which have to be used. It is believed that TGFalpha is the major growth factor overproduced in psoriasis, since 50% of transgenic mice which overexpress TGF alpha develop psoriasis. This suggests that a good inhibitor of EGFR signalling could be used as an antipsoriatic agent, preferably, but not necessarily, by topical dosing.
  • EGF is a potent mitogen for renal tubule cells.
  • Fourfold increases in both EGF urinary secretion and EGF mRNA have been noted in mice with early stage streptozoicin-induced diabetes.
  • increased expression of the EGFR has been noted in patients with proliferative glomerulonephritis (Roychaudhury et al. Pathology 1993, 25, 327).
  • the compounds of the current invention should be useful in treating both proliferative glomerulonephritis and diabetes-induced renal disease.
  • pancreatitis in patients has been reported to correlate with large increases in expression for both EGFR and TGF alpha. (Korc et al. Gut 1994, 35, 1468). In patients showing a more severe form of the disease, typified by an enlargement of the head of the pancreas, there was also shown to be overexpression of the erb-B2 receptor (Friess et al. Ann. Surg. 1994, 220, 183).
  • the compounds of the current invention should prove useful in the treatment of pancreatitis.
  • PCT published application No. WO92/20642 published Nov. 26, 1992 discloses bismono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase.
  • bicyclic pyrimidine derivatives in particular fused heterocyclic pyrimidine derivatives, that are useful at low dosages as inhibitors of EGF-induced mitogenesis. This therefore leads to a further object of compounds having extremely low cytotoxicity.
  • bicyclic pyrimidine derivatives in particular fused heterocyclic pyrimidine derivatives, that have utility as therapeutic agents against proliferative overgrowth diseases, including but not limited to, synovial pannus invasion in arthritis, vascular restenosis, psoriasis and angiogenesis.
  • the compounds disclosed herein also are useful to treat pancreatitis and kidney disease and as a contraceptive agent.
  • At least one, and as many as three of A-E are nitrogen, with the remaining atom(s) carbon, or any two contiguous positions in A-E taken together can be a single heteroatom, N, O or S, in which case one of the two remaining atoms must be carbon, and the other can be either carbon or nitrogen;
  • n 0, 1, 2
  • R 2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcyclo
  • m 0-3, wherein Ar is phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl;
  • R 3 , R 4, R 5 and R 6 are independently, not present, H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms), carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
  • ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms);
  • lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, lower O-alkylhydroxylamino (1-4 carbon atoms);
  • any two of R 3 -R 6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
  • any lower alkyl group substituent on any of the substituents in R 3 -R 6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
  • any of R 3 -R 6 on a neighboring C atom to one of the N atoms cannot be either OH or SH;
  • R 1 , R 2 , R 3 , R 4, R 5 or R 6 contain chiral centers, or in the case of R 1 create chiral centers on the linking atoms, then all stereoisomers thereof both separately and as racemic and/or diastereoisomeric mixtures are included.
  • R 8 is alkyl of from 1-4 carbon atoms or amino or mono or dilower alkyl (1-4 carbon atoms) amino.
  • the invention is also applicable to the compositions of Formulae I and II with the proviso that at least one of the R 3 -R 6 substituents must be taken singly as a substituent other than hydrogen, halo, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms), and with the proviso that A, B, D and E must all be taken singly as carbon or nitrogen atoms.
  • FIG. 1 is an effect of Examples 6 and 7 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 2 is an effect of Example 8 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 3 is a time course for the inhibition of EGF receptor autophosphorylation in A431 by Example 27;
  • FIG. 4 is an effect of Example 27 on EGF receptor autophosphorylation in A431 cells
  • FIG. 5 is an inhibition of EGF receptor autophosphorylation in A431 human epidermoid carcinoma by Example 40;
  • FIG. 6 is an effect of Example 40 on growth factor-mediated tyrosine phosphorylation in Swiss 3T3;
  • FIG. 7 is an effect of Example 40 on growth factor dependent expression of c-jun mRNA in Swiss 3T3 mouse fibroblasts
  • FIG. 8 is an effect of Example 40 on growth factor mediated expression of p39 c-jun ;
  • FIG. 9 is an effect of Example 59 of EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 10 is an effect of Example 60 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 11 is an effect of Example 61 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 12 is an effect of Example 70 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma
  • FIG. 13 is a chart showing an inhibition of EGF receptor tyrosine kinase by Example 27;
  • FIG. 14 is a graph showing an effect of Example 40 on growth factor-mediated mitogenesis in Swiss 3T3 murine fibroblasts
  • FIG. 15 is a photograph of an NIH 3T3 mouse fibroblast line, transfected with the human EGFR gene showing a normal flattened morphology
  • FIG. 16 is a photograph of the same cell line treated with 100 ng/mL of EGF showing a typical spindly transformed morphology
  • FIG. 17 is a photograph of the same cell line in the presence of both 100 ng/mL of EGF and 5 ⁇ m of Example 27 showing the morphology reverted from the transformed type back to the normal type.
  • a suitable ring structure for groups 1-10 is:
  • Suitable ring structures are:
  • a suitable ring structure for groups 12-16 is:
  • a suitable ring structure for groups 17-21 is:
  • a suitable ring structure for groups 22-31 is:
  • a suitable ring structure for groups 33-36 is
  • a suitable ring structure for groups 37-40 is:
  • the compounds of the present invention are prepared according to a number of alternative reaction sequences.
  • Displacement of the 2-chloro of 2,6-dichloro-3-nitropyridine is carried out by cuprous cyanide in NMP. Displacement of the second chlorine of this nitrile by fluoride at this step can be advantageous. This is followed by a mild reduction of the nitro group, under conditions where the halogen is not hydrogenolysed. Hydrolysis of the nitrile followed by orthoformate cyclization, and Vilsmeier-type chlorination will give the dihalopyridopyrimidine. Displacement of the more reactive 4-chlorine with an appropriate amine is followed by displacement of the 6-halogen with the appropriate nucleophile, ammonia, lower alkylamine, hydrazine, methoxide, to form the final products. (NMP is a solvent, N-methyl-2-pyrrolidone).
  • Displacement of chlorine from 2-chloro-3,5-dinitropyridine is accomplished with CUCN in NMP. Reduction of the nitro groups to amines is followed by hydrolysis of the nitrile to an amide. This is cyclized to the pyrimidone with orthoformate, which is converted to the chloride by POCl 3 or possibly turned into the thiomethyl derivative by treatment with phosphorus pentasulfide followed by MeI and a mild base. Displacement with the appropriate amine gives the desired 7-amino compound.
  • the amine functionality can be reductively alkylated or activated by diazotisation of the amino group under acidic or basic conditions, followed by a reduction to the hydrazide, or conversion into a lower alkyl ether, or to a halogen followed by a cuprate or Stille coupling by methods familiar to those skilled in the art.
  • the amine can be reductively aminated, or acylated and reduced to form the alkylamino side chain.
  • ethyl orthoformate may be used instead of formamide for the cyclization, and it may be that some cyclizations will require displacement of F by MeS prior to the reaction.
  • the 4-position is activated by chlorination, and the sidechain amine is then introduced.
  • the final displacement can be by alkoxide or amine nucleophiles to generate the various dialkoxy and amino-alkoxy species, and the appropriate use of R can allow the 7-hydroxyl group to be unmasked at the end of the synthesis.
  • LDA means lithium diisopropyl amide).
  • 2,4-Diamino-5-cyanopyridine can be cyclized directly to many 4-benzylaminopyridopyrimidine derivatives by treatment with the benzylamine and formic acid at high temperature.
  • 2,4-diamino-5-cyanopyridine is converted via ethyl orthoformate/acetic anhydride treatment, followed by cyclization with hydrosulfide ion in anhydrous conditions, to give 7-amino-4-thiono-3H-pyrido[4,3-d]pyrimidine.
  • S-Alkylation and displacement with an appropriate amine gives the desired product. If R 4 is not amino, the amine can be acylated, or reductively alkylated.
  • 2,4-diamino-5-cyanopyridine can be hydrolysed to the corresponding amide, and this species can be cyclized to 7-amino-4-oxo-3H-pyrido[4,3-d]pyrimidine with orthoformate.
  • Diazotization of the 7-amine and replacement with fluorine allows for introduction of other amine and alkoxide nucleophiles at the end of the synthesis after the C4 substituent has been introduced in the usual manner.
  • Diazotization and replacement of the amine with bromide allows for Stille couplings at the 7-position.
  • Nitration of 2-methoxynicotinic acid is followed by displacement of the activated methoxy group and cyclization of the pyrimidone ring, possibly all in one step with formamidine, or alternatively in two steps with ammonia followed by cyclization with a formamide equivalent.
  • the carbonyl is converted to the chloride and displaced with the sidechain in the usual fashion, and the nitro group is then selectively reduced to amino.
  • This can be alkylated, acylated or diazotized.
  • the diazo compound can be converted to hydroxy or to the bromide or iodide, and these latter can undergo a Stille coupling to introduce lower alkyl, alkenyl, aryl, etc. at R 3 .
  • This route uses the known metalation and carboxylation of 2,6-difluoropyridine, followed by displacement of the 2-fluoro substituent. Cyclization of the pyrimidone ring with formamide, followed by conversion of the carbonyl into chloride in a normal manner gives a chlorofluoropyridopyrimidine.
  • the ar(alk)ylamino sidechain is introduced by displacement of the more reactive pyrimidine chlorine, and the R 4 substituent is then introduced by fluoride displacement.
  • the introduction of alkyl utilizes displacement of by alkoxide, later ether cleavage to the pyridone, O-triflation and Stille coupling.
  • This scheme relies on the metalation of 2,6-difluoropyridine similarly to scheme 10.
  • the first metalation is used to introduce oxygen, and the second to introduce the carboxylic acid. If required to is force the second metalation to the 5-position the oxygen may be protected as the very bulky TIPS ether, and stronger basis than LDA may be required.
  • Ammonia is introduced at the 2-position under high temperature and pressure, and the pyridone ring is cyclized, and activated at the 4-position in the usual manner and then displaced with the 4-position sidechain.
  • 5-Bromo-2,6-difluoronicotinic acid is prepared from 2,6-difluoropyridine by successive lithiations using LDA.
  • the 5-position is alkylated via a Stille coupling, and the pyrimidone ring is cyclized on in two steps.
  • the 4-substituent is introduced in the usual fashion and the 7-fluoro group is displaced with thiomethoxide.
  • This thioether in turn is displaced by a Grignard agent in the presence of a nickel salt catalyst.
  • organometallic reagents in the Stille and Grignard couplings could lead to alkenyl, alkynyl and aryl substituents at R 3 and R 4 .
  • Compounds of group 32 are specialized cases of preferred groups 27, 29, 30 and 31, where R 3 and R 4 are cyclized together. They can be made using the same routes as those described for these preferred groups with minor modifications. For example, vicinally substituted alkoxy amino compounds can be dealkylated, and the corresponding vicinal amino alcohols can be bisalkylated with an appropriate dihaloalkane. Piperazines can be made by the route shown in Scheme 13, provided that a suitable amine nucleophile is used to displace the 6-chloro substituent instead of an alkoxide.
  • the pterine nucleus is made by well-established procedure.
  • the pterindione intermediate can be O-alkylated, and for it, and the other groups, the pterindione can be converted to the trichloropterin, and selective displacements can be carried out on the halogens in an order appropriate to give the desired compound.
  • 3,H-Thieno[3,2-d]pyrimid-4-one can be made by standard chemistry from commercially available ethyl 3-aminothiophene carboxylate and formamide. Conversion of the carbonyl to chloride by standard techniques followed by displacement with an appropriate amine gives the desired thieno[3,2-d]pyrimidines. If R 4 is not H, an appropriate electrophile, for example nitro for amine based or diazotization derived substituents, or Br for Stille coupled final products, can be introduced either at the stage shown or an earlier stage, and then be converted to R 4 , by reduction and amination for example or by Stille coupling, or other methods known to those skilled in the art.
  • an appropriate electrophile for example nitro for amine based or diazotization derived substituents, or Br for Stille coupled final products, can be introduced either at the stage shown or an earlier stage, and then be converted to R 4 , by reduction and amination for example or by Stille coupling, or other methods known
  • Thieno[2,3-d]pyrimid-4-one is built up by the Gewald synthesis from 2,5-dihydroxy-1,4-dithiane and ethyl cyanoacetate, followed by formamide cyclization. Conversion of the carbonyl to chloride by standard techniques followed by displacement with an appropriate amine gives the desired thieno[2,3-d]pyrimidines.
  • pyrrolo[2,3-d]pyrimidine a pyrimidine ring is cyclized onto the cyano aminopyrrole using known techniques as shown in scheme B above. Activation and displacement of the thiol by the side chain can be preceded or followed by the optional electrophilic substitution of the pyrrole ring.
  • These compounds are potent and selective inhibitors of the human EGF receptor tyrosine kinase and other members of the EGF receptor family, including the ERB-B2, ERB-B3 and ERB-B4 receptor kinases, and are useful for the treatment of proliferative diseases in mammals. These inhibitors prevent mitogenesis in cells where mitogenesis is driven by one or more of this family of receptor kinases.
  • the preferred compounds are not highly cytotoxic and do not show potent growth inhibitory properties, because of their high specificity toward inhibition of the EGFR kinase family, they should have a much cleaner toxicity profile than most anti-cancer and anti-proliferative drugs. Their very different mode of action to current anti-cancer drugs should allow for their use in multiple drug therapies, where synergism with available agents is anticipated.
  • Human EGF receptor tyrosine kinase was isolated from A431 human epidermoid carcinoma cells which overexpress EGF receptor by the following methods. Cells were grown in roller bottles in 50% Delbuco's Modified Eagle and 50% HAM F-12 nutrient media (Gibco) containing 10% fetal calf serum.
  • Monoclonal antibodies raised to the PDGF (platelet-desired growth factor) receptor or phosphotyrosine were from Upstate Biotechnology, Inc., Lake Placid, N.Y.
  • Anti-pp39 jun antibody to the transcription factor c-jun, which is a 39 kDalton phosphoprotein
  • anti-EGF receptor antibodies were from Oncogene Science, Uniondale, N.Y.
  • the lysate was transferred to a microfuge tube (small centrifuge that holds 1-2 ml plastic centrifuge tubes), allowed to sit on ice 15 minutes and centrifuged 5 minutes at 10,000 ⁇ g. The supernatant was transferred to a clean microfuge tube and 5 ⁇ g of antibody was added to designated samples.
  • the tubes were rotated for 2 hours at 40° C. after which 25 ⁇ l of protein A sepharose was added and then rotation continued for at least 2 more hours.
  • the protein A separose was washed 5 times with 50 mM Hepes, pH 7.5, 150 mM NaCl, 10% glycerol and 0.02% sodium azide.
  • the precipitates were resuspended with 30 ⁇ l of Laemlli buffer (Laemmli, NATURE, Vol. 727, pp. 680-685, 1970), heated to 100° C. for 5 minutes and centrifuged to obtain the supernatant.
  • Laemlli buffer Laemmli, NATURE, Vol. 727, pp. 680-685, 1970
  • Whole cell extracts were made by scraping cells grown in the wells of 6 well plates into 0.2 ml of boiling Laemmli buffer. The extract were transferred to a microfuge tube and heated to 100° C. for 5 minutes.
  • the entire supernatant from the immunoprecipitation or 35 ⁇ l of the whole cell extract was loaded onto a polyacrylamide gel (4-20%) and electrophoresis carried out by the method of Laemlli (Laemmli, 1970).
  • Proteins in the gel were electrophoretically transferred to nitrocellulose and the membrane was washed once in 10 mM Tris buffer, pH 7.2, 150 mM NaCl, 0.01% Azide (TNA) and blocked overnight in TNA containing 5% bovine serum albumin and it ovalbumin (blocking buffer).
  • the membrane was blotted for 2 hours with the primary antibody (1 ⁇ g/ml in blocking buffer) and then washed 2 times sequentially in TNA, TNA containing 0.05% Tween-20 and 0.05% Nonidet P-40 (commercially available detergent) and TNA.
  • the membranes were then incubated for 2 hours in blocking buffer containing 0.1 ⁇ Ci/ml of [ 125 I] protein A and then washed again as above.
  • Protein A is a bacterial protein that specifically bonds certain IgG subtypes and is useful in binding to and isolating antibody-antigen complexes.
  • RNAzol-B (trademark of Tel Test Inc. for a kit used to isolate RNA from tissues) and adhered to the protocol described by the manufacturer. Forty to fifty ⁇ g of RNA was loaded onto a 1% agarose gel and electrophoresis carried out for 3-4 hours at 65 volts. The RNA in the gel was transferred by capillary action to a nylon membrane (Hybond-N, Amersham).
  • the 40 mer c-jun probe was end labeled with [ 32 p]ATP using T4 nucleotide kinase (Promega) and purified on a G25 sephadex column according to the procedure recommended by the supplier, Oncogene Science. Hybridization was performed overnight at 65° C. (c-jun is an immediate early transcription factor; it is one of the components of AP-1 while FOS is the second component of AP-1.
  • Swiss 3T3 fibroblasts were grown to 90-100% confluency in 24- well plates (1.7 ⁇ 1.6 cm, flat bottom) and growth arrested in serum-free media for 18 hours. Drug was added to specified wells 2 hours prior to growth factors and then the cells were exposed to either 20 ng/ml EGF, PDGF or bFGF or 10% serum for 24 hours. Two ⁇ Ci of [methyl- 3 H]thymidine was added to each well and incubated for 2 hours at 37° C. The cells were trypsinized and injected into 2 ml of ice cold 15% trichloroacetic acid (TCA).
  • TCA trichloroacetic acid
  • the resulting precipitate was collected on glassfiber filters, washed five times with 2-ml aliquots of ice-cold 15% TCA, dried and placed in scintillation vials along with 10 ml Ready gel (Beckman, Irvine, Calif.). Radioactivity was determined in a Beckman LS 6800 scintillation counter.
  • B104-1-1 NIH-3T3 fibroblasts transfected by the neu oncogene, Stem et al., Science, 234, pp. 321-324 (1987)
  • SK-BR-3 Human breast carcinoma overexpressing erbB-2 and erbB-3
  • MDA-468 Human breast carcinoma overexpressing the EGF receptor
  • the other lanes show the effect of the stated quantities of the named drug on the growth factor-stimulated activity being measured, demonstrating that the compounds of the present invention have potent effects in whole cells, consistent with their ability to inhibit the tyrosine kinase activity of the EGF receptor.
  • Gel of Example 40 detects mRNA for c-jun by hybridization with a specific radiolabelled RNA probe for c-jun.
  • the gel demonstrates that the growth factors EFG, PDGF and b-FGF stimulate c-jun production in Swiss 3T3 cells, and that compound 40 blocks this production for EGF-stimulated cells, but not for PDGF or b-FGF stimulated cells.
  • This gel shows the amount of c-jun induced in Swiss 3T3 cells by the growth factor EGF, PDGF and b-FGF, quantitating with an anti-c-jun-specific monoclonal antibody. It demonstrates the ability of Example 40 to block c-jun expression in Swiss 3T3 when stimulated by EGF, but not when stimulated by PDGF or b-FGF.
  • the compounds described herein can be used in combination with other components to enhance their activity.
  • additional components are anti-neoplastic materials as, doxorubicin, taxol, cis platin, and the like.
  • 6-Fluoro-3-nitropyridine-2-carboxamide A solution of 2-cyano-6-fluoro-3-nitropyridine (1.40 g, 8.39 mmol) in 90% H 2 SO 4 (30 mL) is warmed at 70° C. for 90 min, then cooled, poured onto ice and basified with conc. ammonia. Extraction with EtOAc and workup gives 6-fluoro-3-nitropyridine-2-carboxamide (0.94 g, 61%).
  • 6-Fluoro-3H-pyrido[3,2-d]pyrimid-4-one A solution of 6-fluoro-3-nitropyridine-2-carboxamide (1.50 g, 8.10 mmol) in EtOAc (80 mL) is hydrogenated over 5% Pd-C (0.30 g) at 60 psi for 2 h. After removal of the catalyst by filtration, the solvent is removed under reduced pressure, to give a residue of crude 3-amino-6-fluoropyridine-2-carboxamide which is used directly in the next step. Triethyl orthoformate (60 mL) is added and the mixture is then heated under reflux with vigorous stirring for 18 h.
  • 6-chloro-3-nitropicolinamide A solution of 6-chloro-3-nitropicolinonitrile (1.00 g, 5.45 mmol) in 90% H 2 SO 4 (15 mL) is warmed at 70° C. for 3.5 h, and then poured into ice-water. The mixture is extracted four times with EtOAc and the combined extracts worked up to give 6-chloro-3-nitropicolinamide (0.80 g, 73%).
  • 6-Chloro-3H-pyrido[3,2-d)pyrimidin-4-one A solution of 6-chloro-3-nitropicolinamide (0.30 g, 1.49 mmol) in EtOAc (30 mL) is hydrogenated at 60 psi over 5% Pd-C (0.10 g) for 20 min. After removal of the catalyst by filtration the solution is concentrated to dryness to give 3-amino-6-chloropicolinamide as a yellow oil, which is used directly in the next step. It is dissolved in triethylorthoformate (30 mL) and the mixture is heated under reflux for 18 h.
  • 4,6-Diamino-2-bromo-3-cyanopyridine (12.77 g, 60 mmol) is hydrogenated in THF/MeOH (240 mL, 2:1) containing KOAc (5.9 g, 60 mmol) and 20% Pd/C (0.5 g) at 18 psi at 25° C. for 4 h.
  • the mixture is celite filtered and the solvent is stripped under reduced pressure to give a solid (11.15 g) which is stirred with THF (100 mL) at room temperature for 20 min.
  • the mixture is refiltered and the filtrate is stripped to dryness to give the desired product.
  • the compound is obtained from 2,4-diamino,5-cyanopyridinium acetate (190 mg, 0.98 mmol), formic acid (0.23 g, 4.4 mmol) and thienylmethylamine (1.07 ml, 10 mmol) as described in a previous experimental.
  • the crude product is converted into a dimesylate salt as described previously and recrystallized from Pr i OH to give 7-amino-4-(thien-2-ylmethylamino)pyrido[4,3-d]pyrimidine dimesylate in 19% yield.
  • Isocuinolinic imide 4-Carboxamidonicotinic acid (280 mg, 1.68 mmol) is heated neat at 200° C. for 5 h to yield isoquinolinic imide (177.2 mg, 71%) as a white solid.
  • 1 H NMR (DMSO) ⁇ 11.68 (H, s)), 9.12-9.03 (2H, m), 7.80 (1H, d, J 5.1 Hz).
  • 5-Amino-2-fluoropyridine is prepared by hydrogenation (Pd/C) of 2-fluoro-5-nitropyridine (obtained from from 2-chloro-5-nitropyridine by reaction with KF in MeCN with Ph 4 PBr [J. H. Clark and D. J. Macquarrie, Tetrahedron Lett., 1987, 28, 111-114]. Reaction of the crude amine with t-Boc anhydride gives 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine.
  • 2,6-Difluoronicotinic acid 2,6-Difluoropyridine (7.89 mL, 0.087 mmol) is added dropwise under N 2 at 78° C. to a stirred solution of lithium diisopropylamide (59.0 mL of a 1.5 N solution in cyclohexane, 0.089 mmol) in THF (250 mL). After 2 h at 78° C., a stream of dry CO 2 is passed through the solution and the mixture is diluted with water and washed with EtOAc.
  • N 6 -Benzyladenine is available commercially from the Aldrich Chemical Company, 1001 West Saint Paul Avenue, Milwaukee, Wis. 53233.
  • compositions of the invention can take any of a wide variety of oral and parenteral dosage forms.
  • the dosage forms comprise as the active components an inhibitor as defined previously.
  • Solid form preparations include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • a solid carrier can be one or more substances which may also act as dilutents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material.
  • the carrier is a finely divided solid which is in admixture with the finely divided active compounds.
  • the active compounds are mixed with carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets preferably contain from 5% or 10% to about 70% of active ingredients.
  • Suitable solid carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low melting wax, cocoa butter, and the like.
  • the term “preparation” is intended to include the formulation of the active compounds with encapsulating materials as carrier, providing a capsule in which the active components (with or without other carriers) are surrounded by carrier, which are thus in association with it.
  • cachets are included. Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration.
  • Liquid form preparations include solutions, suspensions, and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection. Liquid preparations can also be formulated in solution in aqueous polyethylene glycol solution.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizing, and thickening agents as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active components in water with viscous material, i.e., natural or synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other well-known suspending agents.
  • the pharmaceutical preparation is in unit dosage form.
  • the preparation may be subdivided into unit doses containing appropriate quantities of inhibitor and other anti-cancer materials individually or as a combination, i.e., in a mixture.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, for example, packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can also be a capsule, cachet, or tablet itself or it can be the appropriate number of any of these in packaged form.
  • the unit dosage form may be a dividable form having an inhibitor in one part and other anti-cancer materials in the other part, such as, a dividable capsule, a dividable package, or a two-part ampoule, vial or the like.
  • the quantity of an inhibitor in unit dosages of preparation may be varied or adjusted from about 0.01 mg/kg to 100.0 mg/kg, preferably 0.03 mg/kg to less than 1.0 mg/kg of inhibitor.
  • compositions preferably are constituted so that they can be administered parenterally or orally.
  • Solutions of the active compounds as free bases and free acids or pharmaceutically acceptable salts can be prepared in water suitable mixed with a surfactant such as hydroxypropylcellulosa.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of the microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, paragens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferred to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions of agents delaying absorption, for example, gelatin.
  • Sterile-injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients, into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and the freeze-drying technique which yields a powder of active ingredients plus an additional desired ingredient from a previously sterile-filtered solution thereof.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • Dosage unit form refers to physically discrete units suitable as unitary dosages for the mammalian subjects to be treated,; each unit containing a predetermined quantity of active materials calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active materials and the particular therapeutic effect to be achieved, and (b) the limitation inherent in the art of compounding such active materials for the treatment of disease in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail.
  • the principal active ingredients are compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in dosage unit form as hereinbefore disclosed.
  • a unit parenteral dosage form can, for example, contain the principal active compound, i.e. an inhibitor, in amounts ranging from about 0.5 to about 100 mg, with from about 0.1 to 50 mg being preferred.
  • the daily parenteral doses for mammalian subjects to be treated ranges from 0.01 mg/kg to 10 mg/kg of the inhibitor.
  • the preferred daily dosage range is 0.1 mg/kg to 1.0 mg/kg.
  • the daily amount may range from 0.01 mg of active compound/kg of mammalian subject to 100 mg/kg, preferably 0.1 to 10 mg/kg of subject.
  • the inhibitor described above may form commonly known, pharmaceutically acceptable salts such as alkali metal and other common basic salts or acid addition salts, etc. References to the base substances are therefore intended to include those common salts known to be substantially equivalent to the parent compound and hydrates thereof.
  • active compounds described herein are capable of further forming both pharmaceutically acceptable acid addition and/or base salts. All of these forms are within the scope of the present invention.
  • Pharmaceutically acceptable acid addition salts of the active compounds include salts derived from nontoxic inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, hydrofluoric, phosphorous, and the like, as well as the salts derived from nontoxic organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc.
  • nontoxic inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, hydrofluoric, phosphorous, and the like
  • nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids,
  • Such salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, trifluoroacetate.
  • salts of amino acids such as arginate and the like and gluconate, galacturonate (see, for example, Berge, S. M. et al, “Pharmaceutical Salts”, JOURNAL OF PHARMACEUTICAL SCIENCE, 66, pp. 1-19 (1977)).
  • the acid addition salts of said basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner.
  • an active compound can be converted to an acidic salt by treating with an aqueous solution of the desired acid, such that the resulting pH is less than 4.
  • the solution can be passed through a C18 cartridge to absorb the compound, washed with copious amounts of water, the compound eluted with a polar organic solvent such as, for example, methanol, acetonitrile, and the like, and isolated by concentrating under reduced pressure followed by lyophilization.
  • the free base form may be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner.
  • the free base forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base for purposes of the present invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium, and the like.
  • suitable amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge, S. M. et al, “Pharmaceutical Salts”, JOURNAL OF PHARMACEUTICAL SCIENCE, 66, pp. 1-19 (1977)).
  • the base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner.
  • an active compound can be converted to a base salt by treating with an aqueous solution of the desired base, such that the resulting pH is greater than 9.
  • the solution can be passed through a C18 cartridge to absorb the compound, washed with copious amounts of water, the compound eluted with a polar organic solvent such as, for example, methanol, acetonitrile and the like, and isolated by concentrating under reduced pressure followed by lyophilization.
  • the free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner.
  • the free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acids for purposes of the present invention.
  • Certain of the compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms.
  • the solvated forms, including hydrated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention.
  • Certain of the compounds of the present invention possess one or more chiral centers and such center may exist in the R(D) or S(L) configuration.
  • the present invention includes all enantiomeric and epimeric forms as well as the appropriate mixtures thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Rheumatology (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Described are compounds and a method of inhibiting epidermal growth factor receptor tyrosine kinase by treating, with an effective inhibiting amount, a mammal, in need thereof, a compound of Formula II:
Figure US20010027197A1-20011004-C00001
where:
one of A or E is nitrogen, with remaining atoms carbon;
X=O, S, NH or NR7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms). Other terms are described in the specification.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This is a Divisional Application of U.S. Ser. No. 09/191,163, filed Nov. 13, 1998, which is a Divisional Application of U.S. Ser. No. 08/811,797, filed Mar. 6, 1997, which is a Divisional Application of U.S. Ser. No. 08/358,351 filed Dec. 23, 1994, now U.S. Pat. No. 5,654,307, which is a continuation-in-part of U.S. application Ser. No. 08/186,735, filed Jan. 25, 1994 and U.S. application Ser. No. 186,745, filed Jan. 24, 1994, both applications now abandoned.[0001]
  • TECHNICAL FIELD
  • The present invention relates to bicyclic heteroaromatic compounds which inhibit the epidermal growth factor receptor and related receptors and, in particular, their tyrosine kinase enzymic activity. [0002]
  • BACKGROUND ART
  • Cancer is generally a disease of the intracellular signalling system, or signal transduction mechanism. Cells receive instructions from many extracellular sources, instructing them to either proliferate or not to proliferate. The purpose of the signal transduction system is to receive these and other signals at the cell surface, get them into the cell, and then pass the signals on to the nucleus, the cytoskeleton, and transport and protein synthesis machinery. The most common cause of cancer is a series of defects, either in these proteins, when they are mutated, or in the regulation of the quantity of the protein in the cell such that it is over or under produced. Most often, there are key lesions in the cell which lead to a constitutive state whereby the cell nucleus receives a signal to proliferate, when this signal is not actually present. This can occur through a variety of mechanisms. Sometimes the cell may start to produce an authentic growth factor for its own receptors when it should not, the so-called autocrine loop mechanism. Mutations to the cell surface receptors, which usually signal into the cell by means of tyrosine kinases, can lead to activation of the kinase in the absence of ligand, and passing of a signal which is not really there. Alternatively, many surface kinases can be overexpressed on the cell surface leading to an inappropriately strong response to a weak signal. There are many levels inside the cell at which mutation or overexpression can lead to the same spurious signal arising in the cell, and there are many other kinds of signalling defect involved in cancer. This invention touches upon cancers which are driven by the three mechanisms just described, and which involve cell surface receptors of the epidermal growth factor receptor tyrosine kinase family (EGFR). This family consists of the EGF receptor (also known as Erb-B1), the Erb-B2 receptor, and its constituitively active oncoprotein mutant Neu, the Erb-B3 receptor and the Erb-B4 receptor. Additionally, other biological processes driven through members of the EGF family of receptors can also be treated by compounds of the invention described below. [0003]
  • The EGFR has as its two most important ligands Epidermal Growth Factor (EGF) and Transforming Growth Factor alpha (TGFalpha). The receptors appear to have only minor functions in adult humans, but are apparently implicated in the disease process of a large portion of all cancers, especially colon and breast cancer. The closely related Erb-B2 Erb-B3 and Erb-B4 receptors have a family of Heregulins as their major ligands, and receptor overexpression and mutation have been unequivocally demonstrated as the major risk factor in poor prognosis breast cancer. Additionally, it has been demonstrated that all four of the members of this family of receptors can form heterodimeric signalling complexes with other members of the family, and that this can lead to synergistic transforming capacity if more than one member of the family is overexpressed in a malignancy. Overexpression of more than one family member has been shown to be relatively common in human malignancies. [0004]
  • The proliferative skin disease psoriasis has no good cure at present. It is often treated by anticancer agents such as methotrexate, which have very serious side effects, and which are not very effective at the toxicity-limited doses which have to be used. It is believed that TGFalpha is the major growth factor overproduced in psoriasis, since 50% of transgenic mice which overexpress TGF alpha develop psoriasis. This suggests that a good inhibitor of EGFR signalling could be used as an antipsoriatic agent, preferably, but not necessarily, by topical dosing. [0005]
  • EGF is a potent mitogen for renal tubule cells. Fourfold increases in both EGF urinary secretion and EGF mRNA have been noted in mice with early stage streptozoicin-induced diabetes. In addition increased expression of the EGFR has been noted in patients with proliferative glomerulonephritis (Roychaudhury et al. [0006] Pathology 1993, 25, 327). The compounds of the current invention should be useful in treating both proliferative glomerulonephritis and diabetes-induced renal disease.
  • Chronic pancreatitis in patients has been reported to correlate with large increases in expression for both EGFR and TGF alpha. (Korc et al. [0007] Gut 1994, 35, 1468). In patients showing a more severe form of the disease, typified by an enlargement of the head of the pancreas, there was also shown to be overexpression of the erb-B2 receptor (Friess et al. Ann. Surg. 1994, 220, 183). The compounds of the current invention should prove useful in the treatment of pancreatitis.
  • In the processes of blastocyte maturation, blastocyte implantation into the uterine endometrium, and other periimplantation events, uterine tissues produce EGF and TGF alpha (Taga [0008] Nippon Sanka Fujinka Gakkai Zasshi 1992, 44, 939), have elevated levels of EGFR (Brown et al. Endocrinology, 1989, 124, 2882), and may well be induced to produce heparin-binding EGF by the proximity of the developing, but not arrested, blastocyte (Das et al. Development 1994, 120, 1071). In turn the blastocyte has quite a high level of TGF alpha and EGFR expression (Adamson Mol. Reprod. Dev. 1990, 27, 16). Surgical removal of the submandibular glands, the major site of EGF secretion in the body, and treatment with anti-EGFR monoclonal antibodies both greatly reduce fertility in mice (Tsutsumi et al. J. Endocrinology 1993, 138, 437), by reducing successful blastocyte implantation. Therefore, compounds of the current invention should prove to have useful contraceptive properties.
  • PCT patent application Nos. W[0009] 092/07844 published May 14, 1992 and WO92/14716 published September 3, 1992 describe 2,4-diaminoquinazoline as potentiators of chemotherapeutic agents in the treatment of cancer.
  • PCT published application No. WO92/20642 published Nov. 26, 1992 discloses bismono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase. [0010]
  • It is an object of the present invention to inhibit the mitogenic effects of epidermal growth factor utilizing an effective amount of bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives. [0011]
  • It is another object of the present invention to describe bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives, as inhibitors of the EGF, Erb-B2 and Erb-B4 receptor tyrosine kinases. [0012]
  • It is yet another object of the present invention to describe bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives, that are useful at low dosages as inhibitors of EGF-induced mitogenesis. This therefore leads to a further object of compounds having extremely low cytotoxicity. [0013]
  • It is a further object of the present invention to describe bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives, that are useful in suppressing tumors, especially breast cancers, where mitogenesis is heavily driven by EGFR family members. [0014]
  • It is another object of the present invention to describe bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives, that have utility as chronic therapy as inhibitors of EGF-induced responses. [0015]
  • It is another object of the current invention to describe bicyclic pyrimidine derivatives, in particular fused heterocyclic pyrimidine derivatives, that have utility as therapeutic agents against proliferative overgrowth diseases, including but not limited to, synovial pannus invasion in arthritis, vascular restenosis, psoriasis and angiogenesis. The compounds disclosed herein also are useful to treat pancreatitis and kidney disease and as a contraceptive agent. [0016]
  • SUMMARY OF THE INVENTION
  • Described is a method to inhibit epidermal growth factor by treating, with an effective inhibiting amount, a mammal, in need thereof, a compound of Formula I: [0017]
    Figure US20010027197A1-20011004-C00002
  • wherein [0018]
  • at least one, and as many as three of A-E are nitrogen, with the remaining atom(s) carbon, or any two contiguous positions in A-E taken together can be a single heteroatom, N, O or S, in which case one of the two remaining atoms must be carbon, and the other can be either carbon or nitrogen; [0019]
  • X=O, S, NH or NR[0020] 7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms);
  • n=0, 1, 2 [0021]
  • R[0022] 1=H or lower alkyl (1-4 carbon atoms); if n=2, R1 can be independently H or lower alkyl (1-4 carbon atoms) on either linking carbon atom;
  • R[0023] 2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcycloalkyl (3-8 carbon atoms), sulfonylcycloalkyl (3-8 carbon atoms), sulfonamido, lower mono or dialkylsulfonamido (1-4 carbon atoms), mono or dicycloalkylsulfonamido (3-8 carbon atoms), mercapto, carboxy, carboxamido (—C(O)—NH2), lower mono or dialkylcarboxamido (1-4 carbon atoms), mono or dicycloalkylcarboxamido (3-8 carbon atoms), lower alkoxycarbonyl (1-4 carbon atoms), cycloalkoxycarbonyl (3-8 carbon atoms), lower alkenyl (2-4 carbon atoms), cycloalkenyl (4-8 carbon atoms), lower alkynyl (2-4 carbon atoms), or two R2 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring; and
  • m=0-3, wherein Ar is phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl; [0024]
  • R[0025] 3, R4, R5 and R6 are independently, not present, H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms), carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
  • or ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms); [0026]
  • lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, lower O-alkylhydroxylamino (1-4 carbon atoms); [0027]
  • or any two of R[0028] 3-R6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
  • any lower alkyl group substituent on any of the substituents in R[0029] 3-R6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
  • if one or more of A through E are N, then any of R[0030] 3-R6 on a neighboring C atom to one of the N atoms, cannot be either OH or SH; and
  • if any of the substituents R[0031] 1, R2, R3, R4, R5 or R6 contain chiral centers, or in the case of R1 create chiral centers on the linking atoms, then all stereoisomers thereof both separately and as racemic and/or diastereoisomeric mixtures are included.
  • Described also is a method to inhibit epidermal growth factor by treating, with an effective inhibiting amount, a mammal, in need thereof, a compound of Formula II: [0032]
    Figure US20010027197A1-20011004-C00003
  • wherein [0033]
  • Ar, n, m, R[0034] 1-R7 and X are the same as in Formula I;
  • R[0035] 8 is alkyl of from 1-4 carbon atoms or amino or mono or dilower alkyl (1-4 carbon atoms) amino.
  • The invention is also applicable to the compositions of Formulae I and II with the proviso that at least one of the R[0036] 3-R6 substituents must be taken singly as a substituent other than hydrogen, halo, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms), and with the proviso that A, B, D and E must all be taken singly as carbon or nitrogen atoms.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is an effect of Examples 6 and 7 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0037]
  • FIG. 2 is an effect of Example 8 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0038]
  • FIG. 3 is a time course for the inhibition of EGF receptor autophosphorylation in A431 by Example 27; [0039]
  • FIG. 4 is an effect of Example 27 on EGF receptor autophosphorylation in A431 cells; [0040]
  • FIG. 5 is an inhibition of EGF receptor autophosphorylation in A431 human epidermoid carcinoma by Example 40; [0041]
  • FIG. 6 is an effect of Example 40 on growth factor-mediated tyrosine phosphorylation in Swiss 3T3; [0042]
  • FIG. 7 is an effect of Example 40 on growth factor dependent expression of c-jun mRNA in Swiss 3T3 mouse fibroblasts; [0043]
  • FIG. 8 is an effect of Example 40 on growth factor mediated expression of p39[0044] c-jun;
  • FIG. 9 is an effect of Example 59 of EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0045]
  • FIG. 10 is an effect of Example 60 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0046]
  • FIG. 11 is an effect of Example 61 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0047]
  • FIG. 12 is an effect of Example 70 on EGF receptor autophosphorylation in A431 human epidermoid carcinoma; [0048]
  • FIG. 13 is a chart showing an inhibition of EGF receptor tyrosine kinase by Example 27; [0049]
  • FIG. 14 is a graph showing an effect of Example 40 on growth factor-mediated mitogenesis in Swiss 3T3 murine fibroblasts; [0050]
  • FIG. 15 is a photograph of an NIH 3T3 mouse fibroblast line, transfected with the human EGFR gene showing a normal flattened morphology; [0051]
  • FIG. 16 is a photograph of the same cell line treated with 100 ng/mL of EGF showing a typical spindly transformed morphology; and [0052]
  • FIG. 17 is a photograph of the same cell line in the presence of both 100 ng/mL of EGF and 5 μm of Example 27 showing the morphology reverted from the transformed type back to the normal type.[0053]
  • DESCRIPTION OF PREFERRED EMBODIMENTS
  • 1. A preferred form of the invention has X=NH, n=0 or 1, in which case R[0054] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower alkoxy or halogen.
  • 2. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0055] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one amino.
  • 3. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0056] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
  • 4. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0057] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one hydrazino.
  • 5. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0058] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower alkyl.
  • 6. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0059] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 and R4 lower alkoxy.
  • 7. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0060] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 and R4 lower alkyl.
  • 8. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0061] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
  • 9. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0062] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
  • 10. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0063] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 lower mono or dialkylamino, with R4 hydroxy.
  • A suitable ring structure for groups 1-10 is: [0064]
    Figure US20010027197A1-20011004-C00004
  • 11. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0065] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine. Suitable ring structures are:
    Figure US20010027197A1-20011004-C00005
  • 12. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0066] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 lower alkoxy or halogen.
  • 13. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0067] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 amino.
  • 14. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0068] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 lower mono or dialkylamino.
  • 15. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0069] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 hydrazino.
  • 16. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0070] 1=H, the aromatic ring phenyl optionally substituted, A, C & E carbon, with B nitrogen and R4 lower alkyl.
  • A suitable ring structure for groups 12-16 is: [0071]
    Figure US20010027197A1-20011004-C00006
  • 17. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0072] 1 H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 lower alkoxy or halogen.
  • 18. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0073] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 amino.
  • 19. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0074] 1=H, the aromatic ring phenyl optionally substituted, A, B & carbon, with D nitrogen and R3 lower mono or dialkylamino.
  • 20. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0075] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 hydrazino.
  • 21. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0076] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 lower alkyl.
  • A suitable ring structure for groups 17-21 is: [0077]
    Figure US20010027197A1-20011004-C00007
  • 22. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0078] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3or R4 H, with the other one lower alkoxy.
  • 23. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0079] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3or R4 H, with the other one amino.
  • 24. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0080] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
  • 25. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0081] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one hydrazino.
  • 26. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0082] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower alkyl.
  • 27. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0083] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkoxy.
  • 28. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0084] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkyl.
  • 29. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0085] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
  • 30. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0086] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
  • 31. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0087] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R4 lower mono or dialkylamino, with R3 hydroxy.
  • A suitable ring structure for groups 22-31 is: [0088]
    Figure US20010027197A1-20011004-C00008
  • 32. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0089] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
  • 33. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0090] 4 lower alkoxy.
  • 34. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0091] 4 lower mono or dialkylamino.
  • 35. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0092] 4 amino.
  • 36. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0093] 4 hydrazino.
  • A suitable ring structure for groups 33-36 is [0094]
    Figure US20010027197A1-20011004-C00009
  • 37. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0095] 3 and R4 lower alkoxy.
  • 38. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0096] 3 and R4 lower mono or dialkylamino.
  • 39. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0097] 3 or R4 lower alkoxy, with the other lower mono or dialkylamino.
  • 40. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0098] 3 and R4 taken together are ethylenedioxy, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
  • A suitable ring structure for groups 37-40 is: [0099]
    Figure US20010027197A1-20011004-C00010
  • 41. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0100] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom, with D & E carbon, or A & B are carbon with D and E taken together as a sulfur atom, with R4or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 42. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0101] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom, with D & E carbon, or A & B are carbon with D and E taken together as an oxygen atom, with R4 or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 43. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0102] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a nitrogen atom, with D & E carbon, or A & B are carbon with D and E taken together as a nitrogen atom, with R4 or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 44. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0103] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom with D carbon and E nitrogen, or D and E taken together are a sulfur atom, and A is nitrogen and B is carbon, with R3/4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 45. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0104] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom with D carbon and E nitrogen, or D and E taken together are an oxygen atom, and A is nitrogen and B is carbon, with R3/4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 46. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0105] 1=H, the aromatic ring phenyl optionally substituted, A and B taken together are a nitrogen atom, and D is carbon and E is nitrogen, with R3/6 H, or lower alkyl, and R4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 47. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0106] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D nitrogen and E oxygen, with R3/6 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 48. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0107] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D nitrogen and E sulfur, with R3/6 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • 49. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0108] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a nitrogen atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D and E nitrogen atoms, with R3/6 H or lower alkyl if on nitrogen, or H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino if on carbon.
  • Other suitable ring structures are: [0109]
    Figure US20010027197A1-20011004-C00011
  • The compounds of the present invention are prepared according to a number of alternative reaction sequences. [0110]
  • Preparative Routes to Compounds of the Invention
  • [0111] Scheme 1
  • Route for Preferred Groups 1-5, R[0112] 4=H
  • Displacement of the 2-chloro of 2,6-dichloro-3-nitropyridine is carried out by cuprous cyanide in NMP. Displacement of the second chlorine of this nitrile by fluoride at this step can be advantageous. This is followed by a mild reduction of the nitro group, under conditions where the halogen is not hydrogenolysed. Hydrolysis of the nitrile followed by orthoformate cyclization, and Vilsmeier-type chlorination will give the dihalopyridopyrimidine. Displacement of the more reactive 4-chlorine with an appropriate amine is followed by displacement of the 6-halogen with the appropriate nucleophile, ammonia, lower alkylamine, hydrazine, methoxide, to form the final products. (NMP is a solvent, N-methyl-2-pyrrolidone). [0113]
  • 1. A preferred form of the invention has X=NH, n=0 or 1, in which case R[0114] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower alkoxy or halogen.
  • 2. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0115] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one amino.
  • 3. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0116] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
  • 4. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0117] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one hydrazino.
  • 5. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0118] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower alkyl.
  • [0119] Scheme 2
  • Route to Preferred Groups 1-5, R[0120] 3=H
  • Displacement of chlorine from 2-chloro-3,5-dinitropyridine is accomplished with CUCN in NMP. Reduction of the nitro groups to amines is followed by hydrolysis of the nitrile to an amide. This is cyclized to the pyrimidone with orthoformate, which is converted to the chloride by POCl[0121] 3 or possibly turned into the thiomethyl derivative by treatment with phosphorus pentasulfide followed by MeI and a mild base. Displacement with the appropriate amine gives the desired 7-amino compound. The amine functionality can be reductively alkylated or activated by diazotisation of the amino group under acidic or basic conditions, followed by a reduction to the hydrazide, or conversion into a lower alkyl ether, or to a halogen followed by a cuprate or Stille coupling by methods familiar to those skilled in the art. Alternatively, the amine can be reductively aminated, or acylated and reduced to form the alkylamino side chain.
  • [0122] Scheme 3
  • Route to [0123] Preferred Groups 6 and 8-10 where R4=RO
  • The known metalation of 2,6-difluoropyridine is exploited twice. LDA treatment followed by a borate/hydrogen peroxide introduces the 3-hydroxy substituent. If the pyridine undergoes the 2nd metalation at the 4 position, the alcohol can be protected as a TIPS (triisopropyl silyl) ether, which will force the second metalation to the 5-position. Alternative nitrations may be used, such as converting the lithium intermediate to a stannane and treatment with tetranitromethane, or the use of NO[0124] 2BF4 (nitronium tetrafluoroborate). The C1 displacement may be effected by cuprous cyanide or other sources of cyanide ion. After nitrile hydrolysis and nitro group reduction, ethyl orthoformate may be used instead of formamide for the cyclization, and it may be that some cyclizations will require displacement of F by MeS prior to the reaction. The 4-position is activated by chlorination, and the sidechain amine is then introduced. The final displacement can be by alkoxide or amine nucleophiles to generate the various dialkoxy and amino-alkoxy species, and the appropriate use of R can allow the 7-hydroxyl group to be unmasked at the end of the synthesis. (LDA means lithium diisopropyl amide).
  • 6. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0125] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 and R4 lower alkoxy.
  • 8. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0126] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
  • 9. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0127] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
  • 10. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0128] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 lower mono or dialkylamino, with R4 hydroxy.
  • [0129] Scheme 4—Route to Preferred Group 7
  • Use of the 6-alkylquinaldic acid followed by ionic bromination under forcing conditions gives an anhydride, which is opened with ammonia, recyclized to the imide, and then the Hoffman degradation occurs at the less active carbonyl. Cyclization and ring side chain addition in the normal manner is followed by a Stille coupling to introduce the R[0130] 4 alkyl group. At this step alkenyl or aryl substituents could also be introduced using this coupling technology.
  • 7. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0131] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 and R4 lower alkyl.
  • [0132] Scheme 5—Route to Preferred Groups 8, 9, R3=OR
  • Dinitration of 2,6-dihydroxypyridine is followed by conversion to the very reactive dichlorocompound. The dinitrodichloropyridine is singly displaced by cuprous cyanide in NMP, and then the compound is reduced under mild conditions to the diamine. The nitrile is hydrolysed to the amide, which can then be cyclized to the pyridopyrimidone, which is 4-chlorinated in the usual fashion. Displacement of the more reactive chlorine with the 4-sidechain is followed by displacement of the 6-chlorine with alkoxid. For group 9, the amine should be alkylated appropriately by methods familiar to one skilled in the art. [0133]
  • [0134] Scheme 6—Route to Preferred Group 11
  • Compounds of preferred group 11 are specialized cases of [0135] preferred groups 6, 8, 9 and 10, where R3 and R4 are cyclized together. They can be made using the same routes as those described for the preferred groups, with minor modifications, which will be obvious to one skilled in the art. For example vicinally substituted alkoxy amino compounds can be dealkylated, and the corresponding vicinal aminoalcohols can be bisalkylated with an appropriate dihaloalkane.
  • 11. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0136] 1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
  • [0137] Scheme 7—Route for Preferred Groups 12-16
  • 2,4-Diamino-5-cyanopyridine can be cyclized directly to many 4-benzylaminopyridopyrimidine derivatives by treatment with the benzylamine and formic acid at high temperature. For less [0138] nucleophilic amines 2,4-diamino-5-cyanopyridine is converted via ethyl orthoformate/acetic anhydride treatment, followed by cyclization with hydrosulfide ion in anhydrous conditions, to give 7-amino-4-thiono-3H-pyrido[4,3-d]pyrimidine. S-Alkylation and displacement with an appropriate amine gives the desired product. If R4 is not amino, the amine can be acylated, or reductively alkylated. Alternatively 2,4-diamino-5-cyanopyridine can be hydrolysed to the corresponding amide, and this species can be cyclized to 7-amino-4-oxo-3H-pyrido[4,3-d]pyrimidine with orthoformate. Diazotization of the 7-amine and replacement with fluorine allows for introduction of other amine and alkoxide nucleophiles at the end of the synthesis after the C4 substituent has been introduced in the usual manner. Diazotization and replacement of the amine with bromide allows for Stille couplings at the 7-position.
  • 12. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0139] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 lower alkoxy or halogen.
  • 13. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0140] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 amino.
  • 14. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0141] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 lower mono or dialkylamino.
  • 15. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0142] 1=H, the aromatic ring phenyl optionally substituted, A, D & E carbon, with B nitrogen and R4 hydrazino.
  • 16. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0143] 1=H, the aromatic ring phenyl optionally substituted, A, C & E carbon, with B nitrogen and R4 lower alkyl.
  • [0144] Scheme 8—Route for Preferred Groups 17-21
  • [0145] 2-Chloro-5-nitropyridine is converted to the corresponding 2-fluorocompound by KF in DMSO. Reduction of the nitro group followed by treatment with Boc anhydride gives the Bocamino derivative, which can be metalated and carboxylated at the 4-position. Removal of the Boc with TFA and cyclization of the pyrimidone ring with formamide gives 6-fluoro-4-oxo-3H-pyrido[3,4-d]pyrimidine. This is 4-chlorinated in the usual manner and the 4-sidechain is introduced via displacement with an appropriate amine. Displacement of the 6-fluorine with appropriate nucleophiles leads to various different final products. If the fluorine is displaced by thiomethoxide, that in turn can be displaced by alkyl groups in Ni-catalyzed Grignard displacements.
  • 17. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0146] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 lower alkoxy or halogen.
  • 18. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0147] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 amino.
  • 19. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0148] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 lower mono or dialkylamino.
  • 20. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0149] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 hydrazino.
  • 21. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0150] 1=H, the aromatic ring phenyl optionally substituted, A, B & E carbon, with D nitrogen and R3 lower alkyl.
  • Scheme 9—Route to Preferred Groups 22-26, R[0151] 4=H
  • Nitration of 2-methoxynicotinic acid is followed by displacement of the activated methoxy group and cyclization of the pyrimidone ring, possibly all in one step with formamidine, or alternatively in two steps with ammonia followed by cyclization with a formamide equivalent. The carbonyl is converted to the chloride and displaced with the sidechain in the usual fashion, and the nitro group is then selectively reduced to amino. This can be alkylated, acylated or diazotized. The diazo compound can be converted to hydroxy or to the bromide or iodide, and these latter can undergo a Stille coupling to introduce lower alkyl, alkenyl, aryl, etc. at R[0152] 3.
  • 22. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0153] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower alkoxy.
  • 23. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0154] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one amino.
  • 24. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0155] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
  • 25. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0156] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one hydrazino.
  • 26. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0157] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower alkyl.
  • [0158] Scheme 10—Route to Preferred Groups 22-26, R3=H
  • This route uses the known metalation and carboxylation of 2,6-difluoropyridine, followed by displacement of the 2-fluoro substituent. Cyclization of the pyrimidone ring with formamide, followed by conversion of the carbonyl into chloride in a normal manner gives a chlorofluoropyridopyrimidine. The ar(alk)ylamino sidechain is introduced by displacement of the more reactive pyrimidine chlorine, and the R[0159] 4 substituent is then introduced by fluoride displacement. The introduction of alkyl utilizes displacement of
    Figure US20010027197A1-20011004-P00900
    by alkoxide, later ether cleavage to the pyridone, O-triflation and Stille coupling.
  • Scheme 11 [0160]
  • Route to [0161] Preferred Groups 27 and 29-31, R3=RO
  • This scheme relies on the metalation of 2,6-difluoropyridine similarly to [0162] scheme 10. The first metalation is used to introduce oxygen, and the second to introduce the carboxylic acid. If required to is force the second metalation to the 5-position the oxygen may be protected as the very bulky TIPS ether, and stronger basis than LDA may be required. Ammonia is introduced at the 2-position under high temperature and pressure, and the pyridone ring is cyclized, and activated at the 4-position in the usual manner and then displaced with the 4-position sidechain.
  • Displacement of the 7-fluoro substituent with an appropriate nucleophile, followed by conversions as described in previous schemes finishes the synthesis. [0163]
  • 27. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0164] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkoxy.
  • 29. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0165] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
  • 30. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0166] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
  • 31. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0167] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R4 lower mono or dialkylamino, with R3 hydroxy.
  • Scheme 12—Route to Preferred Group 28 [0168]
  • 5-Bromo-2,6-difluoronicotinic acid is prepared from 2,6-difluoropyridine by successive lithiations using LDA. The 5-position is alkylated via a Stille coupling, and the pyrimidone ring is cyclized on in two steps. The 4-substituent is introduced in the usual fashion and the 7-fluoro group is displaced with thiomethoxide. This thioether in turn is displaced by a Grignard agent in the presence of a nickel salt catalyst. Again use of appropriate organometallic reagents in the Stille and Grignard couplings could lead to alkenyl, alkynyl and aryl substituents at R[0169] 3 and R4.
  • 28. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0170] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkyl.
  • Scheme 13—Route to [0171] Preferred Groups 29 and 30, R4=RO
  • Nitration of the commercially available dichloronicotinic acid is followed by a selective displacement of the more reactive Cl under mild conditions, followed by a more forcing displacement of the other Cl, in the appropriate order. The resulting 6-alkoxy-2-amino-5-nitronicotinic acid is cyclized to the pyrimidone, and the 4-carbonyl is converted to a chloride and displaced in the usual fashion with an appropriate amine to give the 4-amino-7-alkoxy-6-nitropyrido[2,3-d]pyrimidine. Reduction of the nitro group, followed by any desired alkylation or acylation gives the desired compounds. [0172]
  • Scheme 14—Route to Preferred Group 32 [0173]
  • Compounds of group 32 are specialized cases of [0174] preferred groups 27, 29, 30 and 31, where R3 and R4 are cyclized together. They can be made using the same routes as those described for these preferred groups with minor modifications. For example, vicinally substituted alkoxy amino compounds can be dealkylated, and the corresponding vicinal amino alcohols can be bisalkylated with an appropriate dihaloalkane. Piperazines can be made by the route shown in Scheme 13, provided that a suitable amine nucleophile is used to displace the 6-chloro substituent instead of an alkoxide.
  • 32. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0175] 1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
  • Scheme 15—Route to Preferred Groups 33-36 [0176]
  • Reaction of a suitable S-alkylisothiouronium salt with methoxymethylidine malononitrile yields a fully functionalized pyrimide precursor. The initially formed pyrimidine can have the SEt displaced by R[0177] 4 either before or after the nitrile hydrolysis, if displacement or oxidation prove problematic later. Displacement of the SEt group can also be achieved without an oxidation to activate the sulfur. Cyclization of the second pyrimidine ring is followed by activation of the 4-carbonyl by thiation and alkylation. Even if the 7-thio group has not been displaced at this point, introduction of the 4-amino sidechain occurs preferentially.
  • 33. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0178] 4 lower alkoxy.
  • 34. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0179] 4 lower mono or dialkylamino.
  • 35. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0180] 4 amino.
  • 36. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, A & D carbon, with B and E nitrogen and R[0181] 4 hydrazino.
  • Scheme 16—Route to Preferred Groups 37-40 [0182]
  • The pterine nucleus is made by well-established procedure. For group 37, the pterindione intermediate can be O-alkylated, and for it, and the other groups, the pterindione can be converted to the trichloropterin, and selective displacements can be carried out on the halogens in an order appropriate to give the desired compound. [0183]
  • 37. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0184] 3 and R4 lower alkoxy.
  • 38. Another preferred form of the invention has X=NH, n=0O the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0185] 3 and R4 lower mono or dialkylamino.
  • 39. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0186] 3 or R4 lower alkoxy, with the other lower mono or dialkylamino.
  • 40. Another preferred form of the invention has X=NH, n=0, the aromatic ring phenyl optionally substituted, B & D carbon, with A and E nitrogen and R[0187] 3 and R4 taken together are ethylenedioxy, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
  • Scheme 17 [0188]
  • Route to Preferred Groups 41 [3,2-d] Ring Fusion [0189]
  • 3,H-Thieno[3,2-d]pyrimid-4-one can be made by standard chemistry from commercially available ethyl 3-aminothiophene carboxylate and formamide. Conversion of the carbonyl to chloride by standard techniques followed by displacement with an appropriate amine gives the desired thieno[3,2-d]pyrimidines. If R[0190] 4 is not H, an appropriate electrophile, for example nitro for amine based or diazotization derived substituents, or Br for Stille coupled final products, can be introduced either at the stage shown or an earlier stage, and then be converted to R4, by reduction and amination for example or by Stille coupling, or other methods known to those skilled in the art. [This technique follows also for all of the following preferred categories which have the possibility of substitution on R3 or R4, as they are all contain electron rich five membered rings which can be readily manipulated by electrophilic aromatic substitution.] (DMSO is dimethyl sulfoxide).
  • 41. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0191] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom, with D & E carbon, or A & B are carbon with D and E taken together as a sulfur atom, with R4 or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 18 [0192]
  • Route to Preferred Groups 41 [2,3-d] Ring Fusion [0193]
  • Thieno[2,3-d]pyrimid-4-one is built up by the Gewald synthesis from 2,5-dihydroxy-1,4-dithiane and ethyl cyanoacetate, followed by formamide cyclization. Conversion of the carbonyl to chloride by standard techniques followed by displacement with an appropriate amine gives the desired thieno[2,3-d]pyrimidines. [0194]
  • Scheme 19 [0195]
  • Route to Preferred Groups 42 [3,2-d] Ring Fusion [0196]
  • The [3,2-d] ring fusion compounds are obtained from 3-bromofurfural as shown above in Scheme A. Displacement of the bromide by azide, followed by oxidation of the aldehyde sets up the basic aminofuroic acid needed to fuse on the pyrimidine ring. The annulation shown can be used, or by manipulating which acid derivative is actually used, one could use a variety of other ring annulations, and subsequent activations of the 4-position if required. [0197]
  • 42. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0198] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom, with D & E carbon, or A & B are carbon with D and E taken together as an oxygen atom, with R4 or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • [0199] Scheme 20
  • Route to Preferred Groups 42 [2,3-d] Ring Fusion [0200]
  • Reaction of 6-chloro-4-methylthiopyrimidine with LDA followed by DMF gives the corresponding 5-aldehyde, which is treated with the sodium salt of an appropriate glycollate ester, displacing chlorine, and in situ forming the furan ring by intramolecular aldol condensation. Cleavage of the ester and decarboxylation of the unwanted 7-acid functionality may be done in a single reaction with a good nucleophile in a dipolar aprotic solvent at high temperature, or in separate saponification and Cu/quinoline decarboxylation steps. Displacement of the 4-methylthio group by an appropriate amine gives the desired furano[2,3-d]pyrimidines. [0201]
  • Scheme 21 [0202]
  • Route to Preferred Groups 43 [2,3-d] Ring Fusion [0203]
  • To make the pyrrolo[2,3-d]pyrimidine a pyrimidine ring is cyclized onto the cyano aminopyrrole using known techniques as shown in scheme B above. Activation and displacement of the thiol by the side chain can be preceded or followed by the optional electrophilic substitution of the pyrrole ring. [0204]
  • 43. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0205] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a nitrogen atom, with D & E carbon, or A & B are carbon with D and taken together as a nitrogen atom, with R4 or R3 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 22 [0206]
  • Route to Preferred Groups 43 [3,2-d] Ring Fusion [0207]
  • The preparation of the pyrrolo[3,2-d]pyrimidine exploits the known condensation of orthoformate with the acidified 4-methyl group of 6-pyrimidones to form the pyrrolopyrimidine as shown above. The side chain can be put on by standard techniques such as in [0208] Scheme 1, and the R4 substituent can be introduced by standard electrophilic chemistry as described above.
  • Scheme 23 [0209]
  • Route to Preferred Groups 44 [5,4-d] Ring Fusion [0210]
  • Condensation of dithioformic acid with 2-aminomalononitrile in the presence of a dehydrating agent such as PPA gives 5-amino-4-cyanothiazole. Reaction of this with orthoformate, followed by treatment with MeSNa gives a thiazolo[5,4-d]pyrimidine derivative, which on treatment with an appropriate amine give the desired compounds. [0211]
  • 44. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0212] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom with D carbon and E nitrogen, or D and E taken together are a sulfur atom, and A is nitrogen and B is carbon, with R3/4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 24 [0213]
  • Route to Preferred Groups 44 [4,5-d] Ring Fusion [0214]
  • Reaction of N-cyanobismethylthiomethyleneimine with ethyl thioglycollate gives ethyl 2-methylthio-4-aminothiazole-5-carboxamide. Cyclization with formamide or equivalent, followed by desulfurization of the methylthio gives a thiazolopyrimidone, which can be activated by Vilsmeier reagent, and the chloride displaced by the desired amine to give the desired thiazolo[4,5-d]pyrimidine derivatives as shown above. [0215]
  • Scheme 25 [0216]
  • Route to Preferred Groups 45 [5,4-d] Ring Fusion [0217]
  • The known 5-amino-4-cyanooxazole is treated with ethyl orthoformate/acetic anhydride, and is then reacted with MeSNa to give 4-methylthiooxazolo[5,4-d]pyrimidine, which on displacement with the appropriate amine gives the desired oxazolo[5,4-d]pyrimidines as shown above. [0218]
  • 45. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0219] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom with D carbon and E nitrogen, or D and E taken together are an oxygen atom, and A is nitrogen and B is carbon, with R3/4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 26 [0220]
  • Route to Preferred Groups 45 [4,5-d] Ring Fusion [0221]
  • Diazotization of the known 5-amino-4,6-dichloropyrimidine, followed by dilute sulfuric acid treatment give the corresponding 5-hydroxy compound. One of the chlorines is displaced with ammonia, and the oxazole ring is annulated with formic acid or an appropriate equivalent. Displacement of the other chlorine with an appropriate amine gives the desired oxazolo[4,5-d]pyrimidines as shown above [0222]
  • [0223] Scheme 27—Route to Preferred Groups 46
  • These compounds can be made by straightforward displacement of halogen on appropriate 6-chloropurines, by means well documented in the art. R[0224] 3 substituents can be introduced via facile electrophilic substitutions at the activated 8-position of the purine nucleus, followed by the types of transformation discussed in previous examples.
  • 46. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0225] 1=H, the aromatic ring phenyl optionally substituted, A and B taken together are a nitrogen atom, and D is carbon and E is nitrogen, with R3/6 H, or lower alkyl, and R4 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 28 [0226]
  • Route to Preferred Groups 47 [5,4-d] Ring Fusion [0227]
  • Reaction of 6-chloro-4-methylthiopyrimidine with LDA followed by DMF gives the corresponding 5-aldehyde, which is treated with hydroxylamine under mild acidic conditions, and then basic conditions to complete the ring annulation giving 4-methylthioisoxazolo[5,4-d]pyrimidine, which on displacement with an appropriate amine gives the desired isoxazolo[5,4-d]pyrimidine derivatives as shown above. [0228]
  • 47. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0229] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are an oxygen atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D nitrogen and E oxygen, with R3/6 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • [0230] Scheme 29
  • Route to Preferred Groups 47 [4,5-d] Ring Fusion [0231]
  • Reaction of 4,6-dichloro-5-nitropyrimidine with CuCN/NMP gives the 4-nitrile. Reduction of the nitro group to the corresponding amine is followed by diazotization and treatment with dilute sulfuric acid to give the corresponding 5-hydroxy compound. Reaction of this with Me[0232] 3Al/NH4Cl gives the amidine which is oxidatively cyclized to 7-amino-4-chloroisoxazolo[4,5-d]pyrimidine. Removal of the amino functionality by diazotization/hypophosphorus acid is followed by displacement of the 4-chlorine with an appropriate amine to give the desired isoxazolo[4,5-d]pyrimidine derivatives as shown above.
  • [0233] Scheme 30
  • Route to Preferred Groups 48 [5,4-d] Ring Fusion [0234]
  • Reaction of 6-chloro-4-methylthiopyrimidine with LDA followed by DMF gives the corresponding 5-aldehyde, which is treated sequentially with NaSH, NBS and ammonia to complete the ring annulation giving 4-methylthioisothiazolo[5,4d]pyrimidine, which on displacement with an appropriate amine gives the desired isothiazolo[5,4-d]pyrimidine derivatives as shown above. [0235]
  • 48. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0236] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a sulfur atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D nitrogen and E sulfur, with R3/6 H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino.
  • Scheme 31 [0237]
  • Route to Preferred Groups 48 [4,5-d] Ring Fusion [0238]
  • Reaction of 4,6-dichloro-5-nitropyrimidine with CuCN/NMP gives the 4-nitrile. Reduction of the nitro group to the amine is followed by diazotization/thiation to give the corresponding 5-mercapto compound. Reaction of this with Me[0239] 3Al/NH4Cl gives the amidine which is oxidatively cyclized with NBS to 7-amino-4-chloroisothiazolo[4,5d]pyrimidine. Removal of the amino functionality by diazotization/hypophosphorus acid is followed by displacement of the 4-chlorine with an appropriate amine to give the desired isothiazolo4,3-d]pyrimidine derivatives as shown above.
  • Scheme 32 [0240]
  • Route to Preferred Groups 49 [3,4-d] Ring Fusion [0241]
  • Reaction of 6-chloro-4-methylthiopyrimidine with LDA followed by DMF gives the corresponding 5-aldehyde, which is treated with hydrazine to do the ring annulation giving 4-methylthiopyrazolo[3,4-d]pyrimidine, which on displacement with an appropriate amine gives the desired pyrazolo[3,4-d]pyrimidine derivatives as shown above. [0242]
  • 49. Another preferred form of the invention has X=NH, n=0 or 1, in which case R[0243] 1=H, the aromatic ring phenyl optionally substituted, and either A and B taken together are a nitrogen atom with D nitrogen and E carbon, or A and B taken together are a carbon atom with D and E nitrogen atoms, with R3/6 H or lower alkyl if on nitrogen, or H, lower alkyl, lower alkoxy, amino, or lower mono or dialkylamino if on carbon.
  • Scheme 33 [0244]
  • Route to Preferred Groups 49 [4,3-d] Ring Fusion [0245]
  • Nitration of pyrazole-3-carboxylic acid followed by reduction gives 4-aminopyrazole-3-carboxylic acid. This is cyclized to pyrazolo[4,3-d]pyrimid-4-one with formamidine HCl, and replacement of the carbonyl with halide by standard procedures, followed by displacement of the chloride by an appropriate amine yields the desired pyrazolo[4,3-d]pyrimidine, as shown above. [0246]
  • Most Preferred Forms of the Invention [0247]
  • 1. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, B, D & E are carbon, A is nitrogen, and R[0248] 4 is amino.
  • 2. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, B, D & E are carbon, A is nitrogen, and R[0249] 4 is methylamino.
  • 3. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, B, D & E are carbon, A is nitrogen, and R[0250] 4 is dimethylamino.
  • 4. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-nitrophenyl, A, D & E are carbon, B is nitrogen, and R[0251] 4 is amino.
  • 5. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0252] 4 is amino.
  • 6. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 4-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0253] 4 is amino.
  • 7. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-trifluoromethylphenyl, A, D & E are carbon, B is nitrogen, and R[0254] 4 is amino.
  • 8. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0255] 4 is acetylamino.
  • 9. A most preferred form of the invention is one where X=NH, x=1, R[0256] 1=H, the aromatic ring is phenyl, A, D & E are carbon, B is nitrogen.
  • 10. A most preferred form of the invention is one where X=NH, x=1, R[0257] 1=H, the aromatic ring is phenyl, A, D & E are carbon, B is nitrogen, and R4 is acetylamino.
  • 11. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, B & E are carbon, D is nitrogen, R[0258] 3=Cl.
  • 12. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, D is nitrogen, and R[0259] 3 is methoxy.
  • 13. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, D is nitrogen, and R[0260] 3 is methylamino.
  • 14. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, D is nitrogen, and R[0261] 3 is dimethylamino.
  • 15. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, D & E are carbon, and A and B taken together are S. [0262]
  • 16. A most preferred form of the invention is one where X=NH, x=1, R[0263] 1=H, the aromatic ring is phenyl, D & E are carbon, and A and B taken together are S.
  • 17. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A & B are carbon, and D and E taken together are S. [0264]
  • 18. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, B is carbon, and A, and D and E taken together, are nitrogen. [0265]
  • 19. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, B & E are carbon, D is nitrogen, and R[0266] 4 is N-piperinyl.
  • 20. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0267] 4 is fluoro.
  • 21. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-hydroxyphenyl, A, D & E are carbon, B is nitrogen, and R[0268] 4 is amino.
  • 22. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0269] 4 is methylamino.
  • 23. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0270] 4 is dimethylamino.
  • 24. A most preferred form of the invention is one where X=NMe, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0271] 4 is methylamino.
  • 25. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, D & E are carbon, B is nitrogen, and R[0272] 4 is methoxy.
  • 26. A most preferred form of the invention is one where X=NH, x=0, the aromatic ring is 3-bromophenyl, A, B & D are carbon, E is nitrogen, and R[0273] 4 is fluoro.
  • Biology [0274]
  • These compounds are potent and selective inhibitors of the human EGF receptor tyrosine kinase and other members of the EGF receptor family, including the ERB-B2, ERB-B3 and ERB-B4 receptor kinases, and are useful for the treatment of proliferative diseases in mammals. These inhibitors prevent mitogenesis in cells where mitogenesis is driven by one or more of this family of receptor kinases. This can include normal cells, where it is desired to prevent mitogenesis, as exemplified by the cells transformed by overexpression or mutation of this kinase family as exemplified by poor prognosis breast cancer where overexpression of EGFR, ERB-B2 and ERB-B3 or mutation of ERB-B2 to the oncoprotein NEU is a major factor in cellular transformation. As the preferred compounds are not highly cytotoxic and do not show potent growth inhibitory properties, because of their high specificity toward inhibition of the EGFR kinase family, they should have a much cleaner toxicity profile than most anti-cancer and anti-proliferative drugs. Their very different mode of action to current anti-cancer drugs should allow for their use in multiple drug therapies, where synergism with available agents is anticipated. [0275]
  • Compounds of the invention have been shown to be very potent, reversible inhibitors of the EGF receptor tyrosine kinase, by binding with high affinity at the adenosine triphosphate (ATP) binding site of the kinase. These compounds exhibit potent IC[0276] 50s, varying from 10 micromolar to 5 picomolar, for the tyrosine kinase activity of the enzyme, based on an assay examining phosphorylation of a peptide derived from the phosphorylation site of the protein PLCgammal, a known EGFR phosphorylation substrate. This data is shown in Table 1.
  • Biological Data
  • Materials and Methods [0277]
  • Purification of Epidermal Growth Factor Receptor Tyrosine Kinase—Human EGF receptor tyrosine kinase was isolated from A431 human epidermoid carcinoma cells which overexpress EGF receptor by the following methods. Cells were grown in roller bottles in 50% Delbuco's Modified Eagle and 50% HAM F-12 nutrient media (Gibco) containing 10% fetal calf serum. Approximately 10[0278] 9 cells were lysed in two volumes of buffer containing 20 mM 2-(4N-[2-hydroxyethyl]piperazin-1-yl)ethanesulfonic acid (hepes), pH 7.4, 5 mM ethylene glycol bis(2-aminoethyl ether) N,N,N′,N′-tetraacetic acid, 1% Triton X-100, 10% glycerol, 0.1 mM sodium orthovanadate, 5 mM sodium fluoride, 4 mM pyrophosphate, 4 mM benzamide, 1 mM dithiothreitol, 80 μg/mL aprotinin, 40 μg/mL leupeptin and 1 mM phenylmethylsulfonyl fluoride. After centrifugation at 25,000×g for 10 minutes, the supernatant was equilibrated for 2 h at 4° C. with 10 mL of wheat germ agglutinin sepharose that was previously equilibrated with 50 mM Hepes, 10% glycerol, 0.1% Triton X-100 and 150 mM NaCl, pH 7.5, (equilibration buffer). Contaminating proteins were washed from the resin with 1 M NaCl in equilibration buffer, and the enzyme was eluted with 0.5 M N-acetyl-1-D-glucosamine in equilibration buffer, followed by 1 mM urea. The enzyme was eluted with 0.1 mg/ml EGF. The receptor appeared to be homogeneous as assessed by Coomassie blue stained polyacrylamide electrophoretic gels.
  • Determination of IC[0279] 50 values—enzyme assays for IC50 determinations were performed in a total volume of 0.1 mL, containing 25 mM Hepes, pH 7.4, 5 mM MgCl2, 2 mM MnCl 21 50 μM sodium vanadate, 5-10 ng of EGF receptor tyrosine kinase, 200 μM of a substrate peptide, (Ac-Lys-His-Lys-Lys-Leu-Ala-Glu-Gly-Ser-Ala-Tyr472-Glu-Glu-Val—NH2,-derived from the amino acid (Tyr472 has been shown to be one of four tyrosines in PLC (phospholipaseC)-gamma 1 that are phosphorylated by the EGF receptor tyrosine kinase [Wahl, M. I.; Nishibe, S.; Kim, J. W.; Kim, H.; Rhee, S. G.; Carpenter, G., J. Biol. Chem., (1990), 265, 3944-3948.], and peptides derived from the enzyme sequence surrounding this site are excellent substrates for the enzyme.),10 μM ATP containing 1 μCi of [32P]ATP and incubated for ten minutes at room temperature. The reaction was terminated by the addition of 2 mL of 75 mM phosphoric acid and passed through a 2.5 cm phosphocellulose filter disc to bind the peptide. The filter was washed five times with 75 mM phosphoric acid and placed in a vial along with 5 mL of scintillation fluid (Ready gel Beckman).
    TABLE 1
    EGF Receptor Tyrosine Kinase Inhibition
    Example # IC 50
     1 8 μM
     2 3.6 μM
     3 1.1 μM
     4 225 nM
     5 1.9 μM
     6 7.6 nM
     7 3.1 nM
     8 9.6 nM
     9 405 nM
    10 6.1 μM
    11 194 nM
    12 13 nM
    13 250 nM
    14 70 nM
    15 134 nM
    16 3.7 μM
    17 1.55 μM
    18 173 nM
    19 1.8 μM
    20 4.9 μM
    21 1.25 μM
    22 39 nM
    23 840 nM
    24 123 nM
    25 377 nM
    26 241 nM
    27 10 nM
    28 94 nM
    29 262 nM
    30 10 μM
    31 15 nM
    32 4.7 μM
    33 130 pM
    34 91 pM
    35 3.1 nM
    36 29 nM
    37 39 nM
    38 71 nM
    39 590 nM
    40 578 nM
    41 220 nM
    42 226 nM
    43 10 μM
    44 10 μM
    45 2.87 μM
    46 1.42 μM
    47 1.67 μM
    48 1.0 μM
    49 2.5 μM
    50 10 μM
    51 1.95 μM
    52 8 μM
    53 1.8 μM
    54 100 nM
    55 400 nM
    56 110 nM
    57 124 nM
    58 40 nM
    59 2.6 nM
    60 8 pM
    61 6 pM
    62 6.1 μM
    63 6.1 μM
    64 11 nM
    65 5.1 μM
    66 190 nM
    67 6.1 μM
    68 263 nM
    69 7.0 μM
    70 473 nM
    71 11 nM
    72 35 nM
    73 36 nM
    74 11.5 μM
    75 55 nM
    76 10 μM
    77 39 nM
    78 670 nM
    79 6.7 nM
  • Cells [0280]
  • Swiss 3T3 mouse fibroblasts, A431 human epidermoid carcinoma cells, and MCF-7 (Michigan Cancer Foundation mammary carcinoma cells), SK-BR-3 (human mammary carcinoma cells), MDA-MB-231 and MDA-MB-468 (human mammary carcinoma cells) breast carcinomas were obtained from the American Type Culture Collection, Rockville, Md. and maintained as monolayers in DMEM (Dulbecco's modified eagle medium)/F12, 50:50 (Gibco/BRL) containing 10% fetal bovine serum. To obtain conditioned medium, MDA-MB-231 cells were grown to confluency in an 850 cm[0281] 2 roller bottle and the medium-replaced with 50 ml of serum-free medium. After 3 days the conditioned medium was removed, frozen down in aliquots and used as a heregulin source to stimulate erbB-2, 3, 4.
  • Antibodies [0282]
  • Monoclonal antibodies raised to the PDGF (platelet-desired growth factor) receptor or phosphotyrosine were from Upstate Biotechnology, Inc., Lake Placid, N.Y. Anti-pp39[0283] jun (antibody to the transcription factor c-jun, which is a 39 kDalton phosphoprotein) and anti-EGF receptor antibodies were from Oncogene Science, Uniondale, N.Y.
  • Immunorecipitation and Western Blot [0284]
  • Cells were grown to 100% confluency in 100 mm Petrie dishes (Corning). After the cells were treated for 5 minutes with either EGF (epidermal growth factor), PDGF, or bFGF (basic fibroblast growth factor) (20 ng/ml) or 1 ml of conditioned media from MDA-MB-231 cells, the media was removed and the monolayer scraped into 1 ml of ice cold lysis buffer (50 mM Hepes, pH 7.5, 150 mM NaCl, 10% glycerol, 1% triton X-100, 1 mM EDTA, 1 mM EGTA, 10 mM sodium pyrophosphate, 30 mM p-nitrophenyl phosphate, 1 mM orthovanadate, 50 mM sodium fluoride, 1 mM phenylmethylsulfonylfluoride, 10 μg/ml of aprotinin, and 10 μg/ml of leupeptin). The lysate was transferred to a microfuge tube (small centrifuge that holds 1-2 ml plastic centrifuge tubes), allowed to sit on ice 15 minutes and centrifuged 5 minutes at 10,000×g. The supernatant was transferred to a clean microfuge tube and 5 μg of antibody was added to designated samples. The tubes were rotated for 2 hours at 40° C. after which 25 μl of protein A sepharose was added and then rotation continued for at least 2 more hours. The protein A separose was washed 5 times with 50 mM Hepes, pH 7.5, 150 mM NaCl, 10% glycerol and 0.02% sodium azide. The precipitates were resuspended with 30 μl of Laemlli buffer (Laemmli, [0285] NATURE, Vol. 727, pp. 680-685, 1970), heated to 100° C. for 5 minutes and centrifuged to obtain the supernatant. Whole cell extracts were made by scraping cells grown in the wells of 6 well plates into 0.2 ml of boiling Laemmli buffer. The extract were transferred to a microfuge tube and heated to 100° C. for 5 minutes. The entire supernatant from the immunoprecipitation or 35 μl of the whole cell extract was loaded onto a polyacrylamide gel (4-20%) and electrophoresis carried out by the method of Laemlli (Laemmli, 1970). Proteins in the gel were electrophoretically transferred to nitrocellulose and the membrane was washed once in 10 mM Tris buffer, pH 7.2, 150 mM NaCl, 0.01% Azide (TNA) and blocked overnight in TNA containing 5% bovine serum albumin and it ovalbumin (blocking buffer). The membrane was blotted for 2 hours with the primary antibody (1 μg/ml in blocking buffer) and then washed 2 times sequentially in TNA, TNA containing 0.05% Tween-20 and 0.05% Nonidet P-40 (commercially available detergent) and TNA. The membranes were then incubated for 2 hours in blocking buffer containing 0.1 μCi/ml of [125I] protein A and then washed again as above. After the blots were dry they were loaded into a film cassette and exposed to X-AR X-ray film for 1-7 days. Protein A is a bacterial protein that specifically bonds certain IgG subtypes and is useful in binding to and isolating antibody-antigen complexes.
  • Northern Blots [0286]
  • Total cellular RNA was isolated from untreated control or treated Swiss 3T3 cells using RNAzol-B (trademark of Tel Test Inc. for a kit used to isolate RNA from tissues) and adhered to the protocol described by the manufacturer. Forty to fifty μg of RNA was loaded onto a 1% agarose gel and electrophoresis carried out for 3-4 hours at 65 volts. The RNA in the gel was transferred by capillary action to a nylon membrane (Hybond-N, Amersham). The 40 mer c-jun probe was end labeled with [[0287] 32p]ATP using T4 nucleotide kinase (Promega) and purified on a G25 sephadex column according to the procedure recommended by the supplier, Oncogene Science. Hybridization was performed overnight at 65° C. (c-jun is an immediate early transcription factor; it is one of the components of AP-1 while FOS is the second component of AP-1.
  • Growth Factor-Mediated Mitogenesis [0288]
  • Swiss 3T3 fibroblasts were grown to 90-100% confluency in 24- well plates (1.7×1.6 cm, flat bottom) and growth arrested in serum-free media for 18 hours. Drug was added to specified [0289] wells 2 hours prior to growth factors and then the cells were exposed to either 20 ng/ml EGF, PDGF or bFGF or 10% serum for 24 hours. Two μCi of [methyl-3H]thymidine was added to each well and incubated for 2 hours at 37° C. The cells were trypsinized and injected into 2 ml of ice cold 15% trichloroacetic acid (TCA). The resulting precipitate was collected on glassfiber filters, washed five times with 2-ml aliquots of ice-cold 15% TCA, dried and placed in scintillation vials along with 10 ml Ready gel (Beckman, Irvine, Calif.). Radioactivity was determined in a Beckman LS 6800 scintillation counter.
  • Growth Inhibition Assay [0290]
  • Cells (2×10[0291] 4) were seeded in 24-well plates (1.7×1.6 cm, flat bottom) in two mls of medium with or without various concentrations of drug. Plates were incubated for 3 days at 37° in a humidified atmosphere containing 5% CO2 in air. Cell growth was determined by cell count with a Coulter Model AM electronic cell counter (Coulter Electronics, Inc., Hialeah, Fla.).
  • INHIBITION OF EGF-INDUCED AUTOPHOSPHORYLATION IN A431 EPIDERMOID CARCINOMA CELLS AND CONDITIONED MEDIA-INDUCED AUTOPHOSPHORYLATION IN SK-BR-3 BREAST TUMOR CELLS BY COMPOUNDS OF THE CURRENT INVENTION
  • [0292]
    Example # EGFR IC50 nM A431 IC50 nM SKBR-3 IC50 nM
     4 225 >1000   >10,000   
     6 7.6 53 2660 
     7 3.1 20 100
     8 9.6 32  71
    22 39 252  ˜1500 
    27 10 110  ˜800 
    59 2.6 12 <10
    60 0.008 13 <10
    61 0.006 21  39
    70 11 124  <10
    74 55 >1000   >1000 
  • ANTIPROLIFERATIVE PROPERTIES OF TYROSINE KINASE INHIBITORS IC50 (n)
  • [0293]
    Ex 60 Ex 61
    B104-1-1 2100  1000
    SK-BR-3  600  900
    MDA-468 3000 12000
  • B104-1-1—NIH-3T3 fibroblasts transfected by the neu oncogene, Stem et al., [0294] Science, 234, pp. 321-324 (1987)
  • SK-BR-3—Human breast carcinoma overexpressing erbB-2 and erbB-3 [0295]
  • MDA-468—Human breast carcinoma overexpressing the EGF receptor [0296]
  • The above gels, developed as detailed in the experimental section, demonstrate the efficacy of compounds of the current invention at blocking certain EGF-stimulated mitogenic signalling events in whole cells. The numbers to the left of the gels indicated the positions of molecular weight standards in kiloDaltons. The lane labelled control shows the degree of expression of the growth-related signal in the absence of EGF stimulation, whereas the lane labelled EGF (or PDGF or b-FGF) shows the magnitude of the growth factor-stimulated signal. The other lanes show the effect of the stated quantities of the named drug on the growth factor-stimulated activity being measured, demonstrating that the compounds of the present invention have potent effects in whole cells, consistent with their ability to inhibit the tyrosine kinase activity of the EGF receptor. [0297]
  • Gel of Example 40 (FIG. 7) detects mRNA for c-jun by hybridization with a specific radiolabelled RNA probe for c-jun. The gel demonstrates that the growth factors EFG, PDGF and b-FGF stimulate c-jun production in Swiss 3T3 cells, and that [0298] compound 40 blocks this production for EGF-stimulated cells, but not for PDGF or b-FGF stimulated cells.
  • Effect of Example 40 on Growth Factor Mediated Expression of p39[0299] c-jun
  • This gel shows the amount of c-jun induced in Swiss 3T3 cells by the growth factor EGF, PDGF and b-FGF, quantitating with an anti-c-jun-specific monoclonal antibody. It demonstrates the ability of Example 40 to block c-jun expression in Swiss 3T3 when stimulated by EGF, but not when stimulated by PDGF or b-FGF. [0300]
  • It is to be appreciated that the compounds described herein can be used in combination with other components to enhance their activity. Such additional components are anti-neoplastic materials as, doxorubicin, taxol, cis platin, and the like. [0301]
  • It has been found that the compounds described herein may inhibit both the erb-B2 and erb-B4 receptors and therefore have significantly increased clinical activity advantageously in combination with the aforementioned anti-neoplastic agents. [0302]
  • See also the results shown in FIGS. [0303] 1-17.
  • Some preferred structures are as follows: [0304]
    Figure US20010027197A1-20011004-C00012
    Ex # Z
    4 fluorine
    6 NH 2
    7 NHCH 3
    8 —N(CH3)2
    Figure US20010027197A1-20011004-C00013
    Ex # Z R2
    22 NH2 —NO2
    27 NH2 Br
    Figure US20010027197A1-20011004-C00014
    Ex # Z R2
    59 —OCH3 Br
    60 —NHCH3 Br
    61 —N(CH3)2 Br
  • Chemical Experimental
  • Listed below are preferred embodiments wherein all temperatures are in degrees Centigrade and all parts are parts by weight unless otherwise indicated. [0305]
  • EXAMPLE 1 4-Anilinopyrido[3,2-d]pyrimidine mesylate
  • 3H-Pyrido[3,2-d]Pyrrimidin-4-one. A solution of 6-chloro-3-nitropicolinamide (2.00 g, 9.91 mmol) in EtOAc/MeOH (1:1, 100 mL) is hydrogenated over 5% Pd-C (0.40 g) at 60 psi for 6 days, with additions of fresh catalyst after 2 and 4 days. After removal of the catalyst by filtration the solution is concentrated to dryness, to give 3-aminopicolinamide as an orange oil, which is used directly in the next step. The crude product is stirred under reflux with triethyl orthoformate (50 mL) for 42 h, during which time a tan precipitate forms. After cooling, the solid is filtered off, washed well with petroleum ether, and dried under vacuum to give 3H-pyrido[3,2-d]pyrimidin-4-one (1.27 g, 87%), mp 343-345° C. [Price, C. C. and Curtin, D. Y. J. Amer. Chem. Soc. 68, 914, 1946 report mp 346-347° C.]. [0306]
  • 4-Chloropyrido[3,2-d]pyrimidine. A suspension of the above pyrimidinone (1.00 g, 6.80 mmol) in POCl[0307] 3 (30 mL) is heated under reflux for 4 h, and then concentrated to dryness under reduced pressure. The residue is partitioned between CH2Cl2 and saturated NaHCO3 solution, and the organic layer worked up to give 4-chloropyrido[3,2-d]pyrimidine (0.97 g, 86%) as a tan solid, mp 335° C. (dec), which is used without further characterisation.
  • 4-Anilinopyrido[3,2-d]pyrimidine mesylate. A solution of 4-chloropyrido[3,2-d]pyrimidine (84 mg. 0.5 mmol), aniline (56 mg, 0.6 mmol) and triethylamine (62 mg, 0.6 mmol) in EtOH (2 mL) are refluxed under N with stirring for 2 h. The crude reaction mixture is purified on a preparative tlc plate (silica), eluting once with 3% MeOH in CHCl[0308] 3. The major band is extracted, and evaporated to dryness under reduced pressure, and the residual solid is dissolved in acetone, (5 mL), filtered, and methanesulfonic acid (32 μL, 0.5 mmol) is added slowly with swirling. The precipitate is collected by suction filtration, rinsed with acetone and dried in a vacuum oven to give 4-anilinopyrido[3,2d]pyrimidine mesylate (91 mg, 57%) as dull yellow needles. 1H NMR (DMSO) δ 11.75 (1H, slbrs), 9.11 (1H, dd, J=1.5, 4.3 Hz), 8.97 (1H, s), 8.32 (1H, dd, J=1.5, 8.4 Hz), 8.12 (1H, dd, J=4.3, 8.5 Hz), 7.88 (2H, d, J=8.2 Hz), 7.49 (2H, t, J=8.0 Hz), 7.32 (1H, t, J=7.0 Hz), 2.34 (3H, s).
  • EXAMPLE 2 4-Benzylaminopyrido[3,2-d]pyrimidine
  • A solution of freshly prepared 4-chloropyrido[3,2-d]pyrimidine (0.10 g, 0.60 mmol) (prepared as described in the previous experimental) and benzylamine (0.13 mL, 1.20 mmol) in propan-2-ol (15 mL) containing a trace of conc. HCl is warmed at 50° C. for 30 min, and then concentrated to dryness. The residue is partitioned between water and EtOAc, and the organic layer worked up and chromatographed on silica gel. EtOAc elutes foreruns, while MeOH/EtOAc (1:9) elutes 4-(benzylamino)pyrido[3,2-d]pyrimidine (0.11 g, 77%). [0309] 1H NMR (CDCl3) δ 8.67 (1H, s), 6.50 (1H, dd, J=4.3, 1.5 Hz), 8.10 (1H, dd, J=8.5, 1.5 Hz), 7.63 (1H, dd, J=8.8, 4.3 Hz), 7.55 (1H, brs), 7.41-7.29 (5H, m), 4.86 (2H, d, J=5.9 Hz).
  • EXAMPLE 3 4- (3-Bromoanilino)pyrido[3,2-d]pyrimidine
  • Reaction of 4-chloropyrido[3,2d]pyrimidine (prepared as described in a previous experimental) with 3-bromoaniline in propan-2-ol containing a trace of conc. HCl at 50° C. for 30 min, followed by chromatography of the product on silica gel, gives 4-(3-bromophenyl)aminopyrido[3,2d]pyrimidine (87% yield). [0310] 1H NMR (CDCl3) δ 9.19 (1H, brs), 8.83 (1H, s), 8.80 (1H, dd, J=4.3, 1.5 Hz), 8.29 (1H, brs), 8.19 (1H, dd, J=8.5, 1.5 Hz), 7.83 (1H, m), 7.76 (1H, dd, J=8.5, 4.3 Hz), 7.29-7.27 (2H, m).
  • EXAMPLE 4 4-(3-Bromoanilino)-6-fluoropyrido[3,2-d]pyrimidine
  • 2-cyano-6-fluoro-3-nitropyridine. A mixture of 6-chloro-2-cyano-3-nitropyridine [Colbry, N. L.; Elslager, E. F. ; Werbel, L. M.; [0311] J. Het. Chem., 1984, 21, 1521-1525](10.0 g, 0.054 mol) and KF (9.48 g, 0.163 mol) in MeCN (200 mL) is heated under reflux with stirring for 18 h, then poured into water and extracted with EtOAc. The extract is washed with water and worked up, and the residue is chromatographed on silica gel, eluting with EtOAc/petroleum ether (3:7), to give after removal of the solvent under reduced pressure 2-cyano-6-fluoro-3-nitropyridine (7.2 g, 79%). 1H NMR (CDCl3) δ 8.79 (1H, dd, J=9.0, 6.0 Hz) 7.48 (1H, dd, J=9.0, 3.0 Hz).
  • 6-Fluoro-3-nitropyridine-2-carboxamide. A solution of 2-cyano-6-fluoro-3-nitropyridine (1.40 g, 8.39 mmol) in 90% H[0312] 2SO4 (30 mL) is warmed at 70° C. for 90 min, then cooled, poured onto ice and basified with conc. ammonia. Extraction with EtOAc and workup gives 6-fluoro-3-nitropyridine-2-carboxamide (0.94 g, 61%). 1H NMR (CDCl3) δ 8.70 (1H, dd, J=8.9, 6.5 Hz), 8.30, 8.03 (1H, 1H, brs), 7.62 (1H, dd, J=8.9, 2.9 Hz).
  • 6-Fluoro-3H-pyrido[3,2-d]pyrimid-4-one. A solution of 6-fluoro-3-nitropyridine-2-carboxamide (1.50 g, 8.10 mmol) in EtOAc (80 mL) is hydrogenated over 5% Pd-C (0.30 g) at 60 psi for 2 h. After removal of the catalyst by filtration, the solvent is removed under reduced pressure, to give a residue of crude 3-amino-6-fluoropyridine-2-carboxamide which is used directly in the next step. Triethyl orthoformate (60 mL) is added and the mixture is then heated under reflux with vigorous stirring for 18 h. The cooled mixture is diluted with an equal volume of petroleum ether, and the resulting precipitate collected by filtration and is washed well with petroleum ether to give 6-fluoro-3H-pyrido[3,2-d]pyrimid-4-one (1.26 g, 84%) . [0313] 1H NMR (DMSO) δ 12.72 (1H, brs), 8.31 (1H, dd, J=8.6, 7.7 Hz), 8.20 (1H, s), 7.66 (1H, dd, J=8.6, 3.0 Hz).
  • 4-(3-Bromoanilino)-6-fluoropyrido[3,2-d)pyrimidine. A suspension of 6-fluoro-3H-pyrido[3,2-d]pyrimid-4-one (0.20 g, 1.21 mmol) in POCl[0314] 3 (30 mL) is heated under reflux with stirring until homogeneous (2 h), and then for a further 1 h. Excess POCl3 is removed under reduced pressure, and the residue is partitioned between CH2Cl2 and saturated aqueous NaHCO3. Workup of the organic portion gives crude 4-chloro-6-fluoropyrido[3,2d]pyrimidine (100%) as an unstable white solid which is used directly in the next step.
  • A solution of 4-chloro-6-fluoropyrido[3,2-d]pyrimidine (0.20 g, 1.1 mmol) and 3-bromoaniline (0.12 mL, 2.18 mmol) in propan-2-ol (20 mL) containing conc. HCl (1 drop) is heated under reflux for 15 min, then cooled, poured into water and extracted with EtOAc. The extract is worked up, and the residue chromatographed on silica gel, eluting with EtOAc/petroleum ether (1:2)to give after removal of the solvent under reduced pressure 4-(3-bromoanilino)-6-fluoropyrido[3,2-d)pyrimidine (0.18 g, 52%). [0315] 1H NMR (CDCl3) δ 8.82 (1H, s), 8.65 (1H, brs), 8.31 (1H, t, J=7.4 Hz), 8.27 (1H, brs), 7.77 (1H, m) 7.41 (1H, dd, J=8.9, 2.2 Hz), 7.29 (2H, brs).
  • EXAMPLE 5 4-(3-Bromoanilino)-6-chloropyrido[3,2d]pyrimidine
  • 6-chloro-3-nitropicolinamide. A solution of 6-chloro-3-nitropicolinonitrile (1.00 g, 5.45 mmol) in 90% H[0316] 2SO4 (15 mL) is warmed at 70° C. for 3.5 h, and then poured into ice-water. The mixture is extracted four times with EtOAc and the combined extracts worked up to give 6-chloro-3-nitropicolinamide (0.80 g, 73%). 1H NMR (DMSO) δ 8.55 (1H, d, J=8.5 Hz), 8.31, 8.04 (1H, 1H, 2 brs), 7.93 (1H, d, J=8.5 Hz).
  • 6-Chloro-3H-pyrido[3,2-d)pyrimidin-4-one. A solution of 6-chloro-3-nitropicolinamide (0.30 g, 1.49 mmol) in EtOAc (30 mL) is hydrogenated at 60 psi over 5% Pd-C (0.10 g) for 20 min. After removal of the catalyst by filtration the solution is concentrated to dryness to give 3-amino-6-chloropicolinamide as a yellow oil, which is used directly in the next step. It is dissolved in triethylorthoformate (30 mL) and the mixture is heated under reflux for 18 h. Petroleum ether (30 mL) is added to the cooled solution, and the resulting precipitate of crude 6-chloro-3H-pyrido[3,2-d)pyrimidin-4-one (0.27 g, 99%) is filtered off and dried in a vacuum oven. [0317]
  • 4-(3-Bromoanilino)-6-chloropyrido[3,2-d]pyrimidine. A suspension of the above quinazolone (0.20 g, 1.10 mmol) in POCl[0318] 3 (30 mL) is heated under reflux for 3 h, and then concentrated to dryness under reduced pressure. The residue is partitioned between CH2Cl2 and saturated NaHCO3 solution, and the organic portion is worked up to give 4,6-dichloropyrido[3,2-d]pyrimidine (0.16 g, 73%) as a tan solid, which is used directly in the next step. A solution of the crude dichloropyridopyrimidine (0.16 g, 0.80 mmol) and 3-bromoaniline (0.17 mL, 1.60 mmol) in propan-2-ol (25 mL) containing a trace of conc. HCl is warmed at 50° C. for 30 min. The cooled mixture is poured into saturated NaHCO3 and extracted with EtOAc, and the extract is worked up and chromatographed on silica gel. Elution with EtOAc/petroleum ether (1:4) gives 3-bromoaniline, while EtOAc/petroleum ether (1:1) elutes 4-(3-bromoanilino)-6-chloropyrido[3,2-d]pyrimidine (0.17 g, 63%). 1H NMR (CDCl3) δ 8.90 (1H, brs,) 8.84 (1H, s), 8.30 (1H, dd, J=2.1, 2.0 Hz) 8.17 (1H, d, J=8.8 Hz), 7.82-7.78 (1H, m) 7.73 (1H, d, J=8.8 Hz), 7.32-7.29, (2H, m).
  • EXAMPLE 6 4-(3-Bromoanilino)-6-aminopyrido[3,2-d]pyrimidine
  • Reaction of 4-(3-bromoanilino)-6-fluoropyrido[3,2-d)pyrimidine (0.12 g, 0.38 mmol)(described in a previous experimental) with a saturated solution of ammonia in ethanol in a pressure vessel at 100° C. for 18 h gives 6-amino-4-(3-bromoanilino)pyrido[3,2d]pyrimidine, (87 mg, 72%). [0319] 1H NMR (CDCl3) δ 8.76 (1H, brs), 8.64 (1H, s), 8.23 (1H, brs), 7.93 (1H, d, J=9.0 Hz), 7.81 (1H, dt, Jd=7.7 Hz, Jt=1.8 Hz), 7.28-7.22 (2H, m), 7.00 (1H, d, J=9.0 Hz), 4.90 (2H, brs).
  • EXAMPLE 7 4-(3-Bromoanilino)-6-methylaminopyrido[3,2-d]pyrimidine
  • Reaction of 4-(3-bromoanilino)-6-fluoropyrido[3,2-d)pyrimidine (50 mg, 0.16 mmol)(described in a previous experimental) with methylamine hydrochloride (32 mg, 0.47 mmol) and triethylamine (70 μL, 0.55 mmol) in ethanol (10 mL) in a pressure vessel at 100° C. for 18 h gives 6-methylamino-4-(3-bromoanilino)pyrido[3,2d]pyrimidine (43 mg, 81%). [0320] 1H NMR (CDCl3) δ 8.81 (1H, brs), 8.61 (1H, s), 8.19 (1H, t, J=1.8 Hz), 7.86 (1H, d, J=9.1 Hz,), 7.83 (1H, dt, Jd=7.7 Hz, Jt=1.8 Hz), 7.28-7.21 (2H, m), 6.92 (1H, d, J=9.1 Hz), 4.97 (1H, q, J=5.0 Hz), 3.13 (3H, d, J=5.0 Hz).
  • EXAMPLE 8 4-(3-Bromoanilino)-6-dimethylaminopyrido[3,2-d]pyrimidine
  • A mixture of 4-(3-bromoanilino)-6-fluoropyrido[3,2-d)pyrimidine (0.15 g, 0.47 mmol) (described in a previous experimental), dimethylamine hydrochloride (0.11 g, 1.41 mmol) and triethylamine (0.23 mL, 1.64 mmol) in EtOH (15 mL) is heated in a pressure vessel at 100° C. for 18 h. The solvent is removed under reduced pressure, and the residue is partitioned between EtOAc and water. The organic portion is worked up, and the residue chromatographed on silica gel. Elution with EtOAc/petroleum ether (1:1) gives foreruns, while EtOAc elutes off 4-(3-bromoanilino)-6-dimethylaminopyrido[3,2-d]pyrimidine (0.14 g, 86%). [0321] 1H NMR (CDCl3) δ 8.72 (1H, brs), 8.56 (1H, s), 8.17 (1H, t, J=1.9 Hz), 7.85 (1H, d, J=9.3 Hz), 7.77 (1H, dt, Jd=7.5 Hz, Jt=1.9 Hz), 7.27-7.18 (2H, m), 7.08 (1H, d, J=9.3 Hz), 3.21 (6H, s).
  • EXAMPLE 9 4-(3-Bromoanilino)-6-methoxypyrido[3,2-d]pyrimidine
  • 4-(3-Bromoanilino)-6-fluoropyrido(3,2-d]pyrimidine (described in a previous experimental) (0.11 g, 0.34 mmol) is added to a solution of NaOMe (prepared by the addition of Na metal (31 mg, 1.38 mmol) to dry MeOH (15 mL). After heating in a pressure vessel at 90° C. for 3 h, the solution is concentrated to dryness and the residue is partitioned between EtOAc and water. Workup of the organic portion gives 4-(3-bromophenyl)amino-6-methoxypyrido[3,2-d]pyrimidine (92 mg, 82%). [0322] 1H NMR (CDCl3) δ 8.73 (1H, s), 8.66 (1H, brs), 8.18 (1H, m), 8.05 (1H, d, J=8.9 Hz), 7.83-7.80 (1H, m), 7.30-7.24 (2H, m), 7.23 (1H, d, J=8.9 Hz), 4.12 (3H, s).
  • EXAMPLE 10 4-Anilinopyrido[4,3-d]pyrimidine
  • 4-(N-t-Butoxycarbonylamino)pyridine. To a mixture of 4-aminopyridine (2 g, 21.24 mmol), potassium hydroxide (3.57 g, 63.72 mmol), water (10 mL), and 2-methyl-2-propanol (4 mL) on ice is added di-t-butyl-dicarbonate (6.95 g, 31.87 mmol). The resulting biphasic solution is stirred at 25° C. for 1 week, then water (20 mL) is added. The solution is extracted with 1×CH[0323] 2Cl2 and 2×EtOAc. The organic layer is dried (MgSO4) and concentrated under reduced pressure to give 4-(N-t-butoxycarbonylamino)pyridine (4.08 g, 99S). 1H NMR (DMSO) δ 9.84 (1H, s), 8.35 (2H, d, J=6 Hz), 7.44 (2H, d, J=7 Hz), 1.49 (9H, s).
  • 4-(N-t-Butoxycarbonylamino)nicotinic acid. n-Butyl lithium (2.18 M, 24 mL, 52.51 mmol) is added slowly to a solution of 4-(N-t-butoxycarbonylamino)pyridine (4.08 g, 21 mmol) in THF (50 mL, stirred under N[0324] 2 at −78° C. The solution is allowed to warm to 0° C., stirred for 3 h, then cooled again to −78° C. and poured into ether (100 mL) containing dry ice. The solution is warmed to room temperature with constant stirring. Water is added and the mixture is neutralized with acetic acid. The resulting solid is collected by vacuum filtration and dried in a vacuum oven to give 4-(N-t-butoxycarbonylamino)nicotinic acid (2.72 g, 54%) as a brown solid. 1H NMR (DMSO) δ 11.75 (1H, brs), 8.95 (1H, s), 8.50 (1H, d, J=6.0 Hz), 8.20 (1H, d, J=6.0 Hz), 1.49 (9H, s).
  • 4-Amino nicotinic acid. A mixture of 4-(N-t-butoxycarbonylamino)nicotinic acid (2.72 g, 11.4 mmol), TFA (10 mL), and CH[0325] 2Cl2 (20 mL) is stirred at room temperature for 12 h. The volatiles are removed under reduced pressure, and the resulting crude 4-amino nicotinic acid is used directly in the next reaction.
  • 3H-Pyrido[4,3-d]pyrimidin-4-one. Crude 4-amino nicotinic acid (2.72 g, 11.4 mmol) in formamide (20 mL) is heated to 170° C. for 12 h. The volatiles are distilled out under reduced pressure (0.8 mmHg). The residual solid is then purified on a medium pressure silica gel column, eluting with 10% MeOH in CHCl[0326] 3 to give 3H-pyrido[4,3-d]pyrimidin-4-one (780 mg, 47%) as a whitish yellow solid. 1H NMR (DMSO) δ 12.64 (1H, brs), 9.28 (1H, s), 8.83 (1H, d, J=5.5 Hz), 8.30 (1H, s), 7.58 (1H, d, J=5.8 Hz).
  • 3H-Pyrido[4,3-d]pyrimidin-4-thione. Phosphorous pentasulfide (2.59 g, 5.83 mmol) is added to a solution of 3H-pyrido[4,3-d]pyrimidin-4-one (780 mg, 5.3 mmol) in pyridine (5 mL). The mixture is refluxed for 5 h. On cooling a precipitate forms and the supernatent is decanted off. The solid is suspended in water (20 mL) and then filtered to yield 3H-pyrido[4,3-d]pyrimidin-4-thione (676 mg, 78%) as a black solid. [0327] 1H NMR (DMSO) δ 14.53 (1H, brs), 9.65 (1H, s), 8.84 (1H, d, J=7.0 Hz), 8.32 (1H, s), 7.64 (1H, d, J=8.0 Hz).
  • 4-Methylthiopyrido[4,3d]pyrimidine. A mixture of 3H-pyrido[4,3-d]pyrimidin-4-thione (676 mg, 4.14 mmol), triethylamine (1.4 mL, 10.31 mmol), DMSO (4 mL), and iodomethane (0.48 mL, 7.72 mmol) is stirred for 12 h under N[0328] 2 at 25° C. The mixture is poured onto water and extracted with EtOAc. The organic extracts are dried (MgSO4), and the solvent is removed under reduced pressure to yield 4-methylthiopyrido[4,3d]pyrimidine (1.15 g, quant.) as a brown solid. 1H NMR (DMSO) δ 9.52 (1H, s), 9.16 (1H, s), 8.95 (1H, d, J=6 Hz), 7.86 (1H, d, J=8 Hz), 2.75 (1H, s).
  • 4-Anilinopyrido[4,3-d]pyrimidine. A mixture of 4-methylthiopyrido[4,3-d]pyrimidine (174 mg, 0.97 mmol), and aniline (186.2 mg, 1.99 mmol) in EtOH (2 mL) is refluxed under N[0329] 2 for 12 h. Cooling to 0° C. forms a solid which is filtered to yield 4-anilinopyrido-[4,3-d]pyrimidine (34.5 mg, 16%). 1H NMR (DMSO) δ 10.29 (1H, brs), 9.86 (1H, s), 8.82 (1H, d, J=5.8 Hz), 8.72 (1H, s), 7.85 (2H, d, J=7.5 Hz), 7.66 (1H, d, J=5.5 Hz), 7.45 (2H, t, J=8.0 Hz), 7.23 (1H, t, J=7.3 Hz).
  • EXAMPLE 11 4-(3-Bromoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 4-methylthiopyrido[4,3-d]pyrimidine (171 mg, 0.96 mmol), (see previous experimental) and 3-bromoaniline (1 mL) is heated to 100° C. for 2 h. A solid precipitates on cooling and is collected by vacuum filtration and then recrystallized from EtOH to yield 4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (30 mg, 10%). [0330] 1H NMR (DMSO) δ 10.33 (1H, s), 9.86 (1H, s), 8.84 (1H, d, J=5.8 Hz), 8.79 (1H, s), 8.22 (1H, s), 7.89 (1H, d, J=7.2 Hz), 7.69 (1H, d, J=5.8 Hz), 7.40 (2H, dt, Jd=8.0 Hz, Jt=1.5 Hz).
  • EXAMPLE 12 4-(3-Bromoanilino)-7-fluoropyrido[4,3d]pyrimidine
  • 3-Cyano-4,6-diaminopyridine. Crude 2-bromo-3-cyano-4,6-diaminopyridine [W. J. Middleton, U.S. Pat. No. 2,790,806 (Apr. 30, 1957), Du Pont; Chem. Abst. 51:P14829 (1957), see also next experimental] (15.1 g, 0.071 mole) is hydrogenated in THF/MeOH (200 mL, 2:1) containing KOAC (7.0 g, 0.071 mole) and 5% Pd/C (4 g) at 55 p.s.i. and 20° C. for 7 days. Filtration over celite, washing with THF/MeOH and removal of the solvent gives a solid, which is dissolved in dilute HCl and water. Adjustment of the solution pH to 10 (conc. NaOH) and cooling gives 3-cyano-4,6-diaminopyridine (6.58 g, 69%) as a yellow solid, mp 197-198° C. [Metzger, R.; Oberdorfer, J.; Schwager, C.; Thielecke, W.; Boldt, P. Liebigs Ann. Chem. 1980, 946-953 record mp (benzene) 205° C.]. Extraction of the remaining liquor with EtOAc (4×200 mL) gives further product (2.12 g, 22w). [0331] 1H NMR (DMSO) δ 7.91 (1H, s), 6.26, 6.24 (2H, 2H, brs), 5.63 (1H, s).
  • 4,6-Diamino-3-pyridylcarboxamide. 3-Cyano-4,6-diaminopyridine (4.30 g, 0.032 mole) is added to 90% H[0332] 2SO4 (25 mL), then stirred at 60-70° C. for 3 h. The resulting solution is added to cold conc. NaOH (40%) to give a mixture of 4,6-diamino-3-pyridylcarboxamide and inorganic salts. An analytically pure sample is obtained by chromatography on alumina (10-50% MeOH/CHCl3) to give a pale yellow solid. 1H NMR (DMSO) δ 8.15 (1H, s) , 6.91 (2H, brs) 7.7-6.3 (2H, brm), 5.78 (2H, brs), 5.56 (1H, s).
  • 7-Amino-4-oxo-3H-pyrido[4,3-d]pyrimidine. [0333] Crude 4,6-diamino-3-pyridylcarboxamide (9.2 g) is heated in purified (EtO)3CH (distilled from Na, 60 mL) at 170° C. for 1.5 d. After removing the solvent, the residue is dissolved in hot 2 M NaOH, filtered, neutralized (conc. HCl) and cooled to give 7-amino-4-oxo-3H-pyrido[4,3-d]pyrimidine (3.57 g, 69% from the nitrile) as a light brown solid 1H NMR (DMSO) δ 11.79 (1H, brs), 8.74 (1H, s), 7.97 (1H, s), 6.76 (2H, brs), 6.38 (1H, s).
  • 7-Fluoro-4-oxo-3H-pyrido[4,3-d]pyrimidine. A solution of 7-amino-4-oxo-3H-pyrido[4,3-d]pyrimidine (1.00 g, 6.17 mmol) in 60% HBF[0334] 4 (25 mL) at 0° C. is treated with solid NaNO2 (0.85 g, 12.3 mmol, added in portions over 2 h), and is then stirred at 0° C. for a further 1 h and at 20° C. for 30 min. The resulting mixture is ice-cooled, neutralized with saturated aqueous Na2CO3, and extracted with EtOAc (4×100 mL). The extract is washed with water, then filtered through silica gel (EtOAc) to give 7-fluoro-4-oxo-3H-pyrido[4,3-d]pyrimidine (0.48 g, 47%) as a cream solid. 1H NMR (DMSO) δ 12.69 (1H, brs), 9.01 (1H, s), 8.31 (1H, s), 7.34 (1H, s)
  • 4-(3-Bromoanilino)-7-fluoropyrido[4,3-d]pyrimidine. A suspension of 7-fluoro-4-oxo-3H-pyrido[4,3d]pyrimidine (0.23 g, 1.39 mmol) in POCl[0335] 3 (10 mL) is stirred under reflux for 3.5 h, and is then concentrated under vacuum. The resulting oil is ice-cooled, diluted with CH2Cl2 (100 mL), saturated aqueous Na2CO3 (40 mL) and ice, and stirred at 20° C. for 2 h. The CH2Cl2 extract is separated and the aqueous portion further extracted with CH2Cl2 (2×100 mL), and then the combined extracts are dried (Na2SO4) and filtered to give crude 4-chloro-7-fluoropyrido[4,3-d]pyrimidine. 3-Bromoaniline (1.26 g, 7.35 mmole), 3-bromoaniline hydrochloride (20 mg) and dry isopropanol (5 mL) are added, then the resulting solution is concentrated under vacuum to remove the CH2Cl2 and stirred at 20° C. for 1 h. Upon addition of dilute NaHCO3 and water, the product crystallises. Filtration, washing with water and CH2Cl2, gives pure 4-(3-bromoanilino)-7-fluoropyrido[4,3-d]pyrimidine (297 mg, 67%) as a cream solid. 1H NMR (DMSO) δ 10.38 (1H, brs), 9.59 (1H, s), 8.72 (1H, s), 8.17 (1H, s), 7.85 (1H, m) , 7.38 (3H, m).
  • EXAMPLE 13 7-Amino-4-anilinopyrido[4,3-d]pyrimidine
  • 4,6-Diamino-2-bromo-3-cyanopyridine. HBr is bubbled for 2 h into a mixture of malononitrile (16.3 g, 0.247 mol) and toluene (400 mL) at 0° C. A light yellow precipitate forms. The reaction mixture is then heated at 100° C. for 2 h, with much gas evolution. After cooling to room temperature, the yellow solid is isolated via suction filtration, washed with toluene and air dried. The solid (25.96 g) is mixed with water (500 mL), and the pH of the suspension is adjusted to 9˜10 with NH[0336] 4OH (conc. ˜15 mL). After stirring at room temperature for 1 h, the mixture is filtered. Recrystallization from EtOH affords a yellow solid. After drying at 60° C. in a vacuum oven, 4,6-diamino-2-bromo-3-cyanopyridine (12.95 g, 49%) is obtained. 1H NMR (DMSO) δ 6.67 (2H,brs), 6.55 (2H,brs), 5.59 (1H,s). 2,4-Diamino-5-cyanopyridinium acetate. 4,6-Diamino-2-bromo-3-cyanopyridine (12.77 g, 60 mmol) is hydrogenated in THF/MeOH (240 mL, 2:1) containing KOAc (5.9 g, 60 mmol) and 20% Pd/C (0.5 g) at 18 psi at 25° C. for 4 h. The mixture is celite filtered and the solvent is stripped under reduced pressure to give a solid (11.15 g) which is stirred with THF (100 mL) at room temperature for 20 min. The mixture is refiltered and the filtrate is stripped to dryness to give the desired product. After drying in a vacuum oven, 2,4-diamino-5-cyanopyridinium acetate (10.65 g, 92%) is collected as a yellow solid. 1H NMR (DMSO) δ 7.90 (1H, s), 6.26 (4H, brs), 5.62 (1H, s), 1.90 (3H, s).
  • 7-Amino-4-thiono-3H-pyrido[4,3-d]pyrimidine. A mixture of 2,4-diamino-5-cyanopyridinium acetate (0.199 g, 1.0 mmol), triethyl orthoformate (1.95 mL) and Ac[0337] 2O (1.95 mL) is refluxed under N2 with stirring for 3 h. The solvent is then stripped and the residue is dissolved in MeOH (10 mL) containing NaOMe (0.81 g, 15 mmol). H2S is bubbled through the mixture for ˜5 min, which is then refluxed overnight. After the solvent is stripped, the residue is dissolved in hot water and boiled with charcoal. After filtration, the filtrate is neutralized with acetic acid whilst hot to generate a yellow solid. On cooling, the solid is collected by suction filtration, and is dried in a vacuum oven overnight. 7-Amino-4-thiono-3H-pyrido[4,3-d]pyrimidine (84 mg, 51%) is isolated as light yellow solid. 1H NMR (DMSO) δ 9.82 (1H, s) , 9.34 (1H, s), 8.37 (1H, s), 7.80 (2H, d, J=7.5 Hz), 7.38 (2H, t, J=7.5 Hz), 7.12 (1H, t, J=7.5 Hz), 6.61 (2H, brs) 6.43 (1H, s).
  • 7-Amino-4-methylthiopyrido[4,3-d]pyrimidine. NEt[0338] 3 (6 mL, 43 mmol) is added to a solution of 7-amino-4-thiono-3H-pyrido[4,3-d]pyrimidine (0.77 g, 4.3 mmol) in DMSO (7 mL) stirred under N2 at 25° C. After the two phases have been stirred for 20 min, MeI (0.26 mL, 4.2 mmol) is added. After 2 h, the reaction mixture is poured onto stirring ice-water. Solid forms instantly. After further cooling at 0° C., the solid is collected by suction filtration and dried in a vacuum oven to give 7-amino-4-methylthiopyrido[4,3d]pyrimidine (0.564 g, 68%). 1H NMR (DMSO) δ 8.98 (1H, s), 8.71 (1H, s), 6.94 (2H, brs), 6.49 (1H, s) 2.63 (3H, s).
  • 7-Amino-4-anilinopyrido[4,3-d]pyrimidine. A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (0.136 g, 0.7 mmol) and aniline (0.5 mL, 5.5 mmol) is refluxed under N[0339] 2 at 180° C. for 2 h. The reaction mixture is cooled to 25° C., when it precipitates. The solid is collected by suction filtration and recrystallized from isopropanol, and dried in a vacuum oven overnight. 7-Amino-4-anilinopyrido[4,3-d]pyrimidine (84 mg, 51%) is isolated as a light yellow solid. 1H NMR (DMSO) δ 9.82 (1H, s), 9.34 (1H, s), 8.37 (1H, s), 7.80 (2H, d, J=7.5 Hz), 7.38 (2H, t, J=7.5 Hz), 7.12 (1H, t, J=7.5 Hz), 6.61 (2H, brs) 6.43 (1H, s).
  • EXAMPLE 14 7-Amino-4-(3-hydroxyanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (299 mg, 1.56 mmole) and 3-aminophenol (1.60 g, 14.7 mmole) is stirred at 160° C. for 15 min. The resulting product is chromatographed over silica gel (9% MeOH/CH[0340] 2Cl2) to give 7-amino-4-(3-hydroxyanilino)pyrido[4,3-d]pyrimidine (108 mg, 18%) as a pale orange solid. 1H NMR (DMSO) δ 9.69 (1H, brs), 9.44 (1H, brs), 9.33 (1H, s), 8.38 (1H, s), 7.37 (1H, t, J=2.1 Hz), 7.21 (1H, brd, J=8.4 Hz), 7.14 (1H, t, J=8.0 Hz), 6.59 (2H, brs), 6.53 (1H, ddd, J=7.9, 2.2, 0.8 Hz), 6.43 (1H, s).
  • EXAMPLE 15 7-Amino-4-(3-methoxyanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (226 mg, 1.18 mmol) (described in the previous experimental) and m-anisidine (1.00 mL, 8.90 mmol) is stirred under N[0341] 2 at 190° C. for 1.5 h. The resulting product is chromatographed over silica gel (5-7% EtOH/EtOAc) to give 7-amino-4-(3-methoxyanilino)pyrido[4,3d]pyrimidine (136 mg, 43%) as a light brown solid. 1H NMR (DMSO) δ 9.78 (1H, brs), 9.34 (1H, s), 8.40 (1H, s), 7.50 (1H, brs), 7.44 (1H, d, J 8.0 Hz), 7.28 (1H, t, J=8.2 Hz), 6.71 (1H, dd, J=8.2, 2.3 Hz), 6.61 (2H, brs), 6.45 (1H, s), 3.77 (3H, s).
  • EXAMPLE 16 7-Amino-4-(2-methoxyanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (227 mg, 1.18 mmole) and o-anisidine (1.00 mL, 8.87 mmol) is stirred under N[0342] 2 at 180° C. for 2.5 h. The resulting product is chromatographed over silica gel (5% EtOH/EtOAc) to give 7-amino-4-(2-methoxyanilino)pyrido[4,3-d]pyrimidine (147 mg, 47%) as a yellow solid. 1H NMR (DMSO) δ 9.44 (1H, brs) , 9.25 (1H, s), 8.22 (1H, s), 7.54 (1H, dd, J=7.7, 1.4 Hz), 7.24 (1H, ddd, J=8.1, 7.4, 1.5 Hz), 7.10 (1H, dd, J=8.2, 1.2 Hz), 6.98 (1H, dt, Jd=1.3 Hz, Jt=7.5 Hz), 6.52 (2H, brs), 6.41 (1H, s), 3.79 (3H, s).
  • EXAMPLE 17 7-Amino-4-(3-aminoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (307 mg, 1.60 mmol) (described in a previous experimental) and 3-nitroaniline (2.00 g, 14.5 mmol) is stirred at 200° C. for 1.5 h, and the crude product is suspended in MeOH/THF (4:1, 250 mL) and hydrogenated over 5% Pd/C (2 g) at 60 psi and 20° C. for 24 h. The solution is filtered over celite, washing thoroughly (hot MeOH), and is then absorbed onto alumina and chromatographed on alumina (4-8% EtOH/CHCl[0343] 3) to give 7-amino-4-(3-aminoanilino)pyrido[4,3-d]pyrimidine (66 mg, 16%) as a green solid,. 1H NMR (DMSO) δ 9.57 (1H, brs), 9.30 (1H, s), 8.33 (1H, s), 7.04 (1H, t, J=2.0 Hz), 6.99 (1H, t, J=8.0 Hz), 6.88 (1H, brd, J=8.0 Hz), 6.55 (2H, brs), 6.40 (1H, s), 6.34 (1H, dd, J=7.9, 1.3 Hz), 5.10 (2H, brs).
  • EXAMPLE 18 7-Amino-4-(4-aminoanilino)pyrido[4,3-d]pyrimidine
  • 7-Amino-4-(4-acetamidoanilino)pyrido[4,3-d]pyrimidine. A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (138 mg, 0.72 mmole) and 4-aminoacetanilide (1.50 g, 10.0 mmole) is stirred under N[0344] 2 at 200° C. for 1 h. The resulting product is chromatographed over alumina (8-10% MeOH/CH2Cl2) to give 7-amino-4-(4-acetamidoanilino)pyrido[4,3-d]pyrimidine (110 mg, 52%) as a pale yellow solid. 1H NMR (DMSO) δ 9.94, 9.79 (1H, 1H, 2 brs), 9.31 (1H, s), 8.34 (1H, s), 7.69 (2H, d, J=8.9 Hz), 7.57 (2H, d, J=8.9 Hz), 6.57 (2H, brs), 6.43 (1H, s), 2.05 (3H, s).
  • 7-Amino-4-(4-aminoanilino)pyrido[4,3-d]pyrimidine. A solution of 7-amino-4-(4-acetamidoanilino)pyrido[4,3-d]pyrimidine (0.30 g, 1.02 mmole) in aqueous NaOH (2 M, 10 mL) and MeOH (10 mL) is stirred at 100° C. for 7 h. The resulting product is chromatographed over alumina (3-4% EtOH/CHCl[0345] 3) to give 7-amino-4-(4-aminoanilino)pyrido[4,3d]pyrimidine (86 mg, 33%) as an orange solid. 1H NMR (DMSO) δ 9.58 (1H, brs), 9.24 (1H, s), 8.25 (1H, s), 7.31 (2H d, J=8.6 Hz), 6.58 (2H, d, J 8.6 Hz), 6.48 (2H, brs), 6.39 (1H, s), 5.00 (2H, brs).
  • EXAMPLE 19 7-Amino-4- (3-dimethylaminoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (245 mg, 1.28 mmol) (described in a previous experimental) and N,N-dimethyl-1,3-phenylenediamine (1.60 g, 11.8 mmol) is stirred under N[0346] 2 at 190° C. for 1 h, and the resulting product is chromatographed (twice) over alumina (3% EtOH/CHCl3) to give 7-amino-4-(3-dimethylaminoanilino)pyrido[4,3-d]pyrimidine (113 mg, 32%) as a pale yellow solid. 1H NMR (DMSO) δ 9.66 (1H, brs), 9.33 (1H, s), 8.36 (1H, s), 7.22 (1H, brd, J=7.8 Hz), 7.16 (2H, m), 6.57 (2H, brs), 6.51 (1H, ddd, J=8.0, 2.3, 1.2 Hz), 6.42 (1H, s), 2.91 (6H, s).
  • EXAMPLE 20 7-Amino-4-(4-dimethylaminoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (256 mg, 1.33 mmole) and N,N-dimethyl-1,4-phenylenediamine (1.95 g, 14.4 mmole) is stirred under N[0347] 2 at 190° C. for 20 min. The resulting product is chromatographed over alumina (3-7% EtOH/CHCl3) to give 7-amino-4-(4-dimethylaminoanilino)pyrido[4,3-d]pyrimidine (198 mg, 53%) as an orange solid. 1H NMR (DMSO) δ 9.67 (1H, brs), 9.27 (1H, s), 8.27 (1H, s), 7.51 (2H, d, J 8.9 Hz), 6.75 (2H, d, J=8.9 Hz), 6.51 (2H, brs) , 6.39 (1H, s), 2.89 (6H, s)
  • EXAMPLE 21 7-Amino-4-(2-nitroanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (220 mg, 1.15 mmole) and 2-nitroaniline (2.00 g, 14.5 mmole) is heated to 100° C., then excess dry HCl gas is added to the hot stirred solution, and the mixture stirred at 160° C. for 20 min. The resulting product is neutralized with excess NaHCO[0348] 3, dissolved in MeOH/CHCl3, dried onto silica gel and chromatographed over silica gel (2-4% MeOH/CH2Cl2) to give 7-amino-4-(2-nitroanilino)pyrido[4,3-d]pyrimidine (108 mg, 33%) as a yellow brown solid. 1H NMR (DMSO) δ 10.40 (1H, brs), 9.24 (1H, brs), 8.20 (1H, brs), 8.12 (1H, brs), 8.01 (2H, brs), 7.75 (1H, brs), 6.70 (2H, brs), 6.43 (1H, brs).
  • EXAMPLE 22 7-Amino-4- (3-nitroanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (127 mg, 0.66 mmol) (described in a previous experimental) and 3-nitroaniline (1.70 g, 12.3 mmol) is stirred under N[0349] 2 at 200° C. for 1.5 h. The resulting product is chromatographed over alumina (5-20% EtOH/CHCl3) to give 7-amino-4-(3-nitroanilino)pyrido[4,3d]pyrimidine (81 mg, 39%) as a brown solid. 1H NMR (DMSO) δ 10.17 (1H, brs), 9.37 (1H, s), 8.87 (1H, brs), 8.48 (1H, s), 8.33 (1H, brd, J=7.5 Hz), 7.95 (1H, ddd, J=8.2, 2.1, 1.0 Hz), 7.67 (1H, t, J 8.2 Hz), 6.70 (2H, brs), 6.47 (1H, s).
  • EXAMPLE 23 7-Amino-4-(3-fluoroanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (215 mg, 1.12 mmol) and 3-fluoroaniline (1.16 g, 10.4 mmol) is stirred at 160° C. for 30 min. The resulting product is chromatographed over silica gel (6-7% MeOH/CH[0350] 2Cl2) to give 7-amino-4-(3-fluoroanilino)pyrido[4,3-d]pyrimidine (185 mg, 65%) as a white solid. 1H NMR (DMSO) δ 9.94 (1H, brs), 9.36 (1H, s), 8.46 (1H, s), 7.91 (1H, brd, J=11.9 Hz), 7.63 (1H, brd, J=8.1 Hz), 7.41 (1H, dd, J=15.7, 7.7 Hz), 6.93 (1H, dt, Jt=8.5 Hz, Jd=2.4 Hz), 6.68 (2H, brs) , 6.38 (1H, s)
  • EXAMPLE 24 7-Amino-4-(3-chloroanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (208 mg, 1.08 mmol) and 3-chloroaniline (1.21 g, 9.48 mmol) is stirred at 150° C. for 20 min. The resulting product is chromatographed over alumina (5-10% MeOH/CH[0351] 2Cl2) to give 7-amino-4-(3-chloroanilino)pyrido[4,3-d]pyrimidine (177 mg, 60%) as a white solid. 1H NMR (DMSO) δ 9.92 (1H,brs), 9.35 (1H, s), 8.45 (1H, s), 8.08 (1H, brs), 7.79 (1H, brd, J=8.0 Hz), 7.40 (1H, t, J=8.1 Hz), 7.16 (1H, dd, J=7.9, 1.3 Hz), 6.68 (2H, brs), 6.46 (1H, s).
  • EXAMPLE 25 7-Amino-4-(3,4-dichloroanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (247 mg, 1.29 mmol) and 3,4-dichloroaniline (1.50 g, 9.26 mmol) is stirred at 165° C. for 30 min. The resulting product is chromatographed over silica gel (7-8% MeOH/CH[0352] 2Cl2) to give 7-amino-4-(3,4-dichloroanilino)pyrido[4,3-d]pyrimidine (252 mg, 64%) as a pale yellow solid. 1H NMR (DMSO) δ 9.97 (1H, brs), 9.34 (1H, s), 8.47 (1H, s), 8.29 (1H, brs), 7.86 (1H, brd, J=8.6 Hz), 7.62 (1H, d, J=8.8 Hz), 6.70 (2H, brs), 6.46 (1H, s).
  • EXAMPLE 26 7-Amino-4-(2-bromoanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (198 mg, 1.03 mmol) (described in a previous experimental) and 2-bromoaniline (1.00 mL, 9.18 mmol) is stirred under N[0353] 2 at 180° C. for 2.5 h, and the resulting product is chromatographed on alumina (1% EtOH/CHCl3) to give 7-amino-4-(2-bromoanilino)pyrido[4,3d]pyrimidine (108 mg, 33%) as a pale yellow solid,1H NMR (DMSO) δ 9.91 (1H, brs), 9.27 (1H, s), 8.20 (1H, s), 7.73 (1H, d, J=7.9 Hz), 7.50 (1H, m), 7.44 (1H, t, J=6.9 Hz), 7.25 (1H, m), 6.59 (2H, brs), 6.42 (1H, s).
  • EXAMPLE 27 7-Amino-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (167 mg, 0.87 mmol) (described in a previous experimental) and 3-bromoaniline (0.75 mL, 7.8 mmol) is stirred under N[0354] 2 at 190° C. for 2.5 h, and the precipitate which appears on cooling is recrystallized from Pr1OH. 1H NMR (DMSO) δ 9.91 (1H, brs), 9.34 (1H, s), 8.45 (1H, s), 8.19 (1H, s), 7.84 (1H, d, J=8.0 Hz), 7.34 (1H, t, J=8.0 Hz), 7.29 (1H, d, J=8.2 Hz), 6.68 (2H, brs), 6.45 (1H, s).
  • EXAMPLE 28 7-Amino-4-(4-bromoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido [4,3-d]pyrimidine (261 mg, 1.36 mmole) and 4-bromoaniline (1.00 g, 5.81 mmole) is stirred under N[0355] 2 at 200° C. for 15 min. The resulting product is chromatographed on silica gel (10-15% EtOH/EtOAc) to give 7-amino-4-(4-bromoanilino)pyrido[4,3-d]pyrimidine (200 mg, 46%) as a pale yellow solid. 1H NMR (DMSO) δ 9.88 (1H, brs). 9.34 (1H, s), 8.40 (1H, s), 7.83 (2H, d, J=8.8 Hz 7.55 (2H, d, J=8.8 Hz), 6.64 (2H, brs), 6.44 (1H, s).
  • EXAMPLE 29 7-Amino-4-(3-iodoanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (72 mg, 0.37 mmol) and 3-iodoaniline (1.25 g, 5.71 mmol) is stirred at 160° C. for 30 min. The resulting product is chromatographed over silica gel (5-7% MeOH/CH[0356] 2Cl2) to give 7-amino-4-(3-iodoanilino)pyrido[4,3d]pyrimidine (83 mg, 61%) as a light brown rosettes. 1H NMR (DMSO) δ 9.84 (1H, brs), 9.34 (1H, s), 8.44 (1H, s), 8.30 (1H, brs), 7.90 (1H, dd, J=7.9, 0.8 Hz), 7.47 (1H, d, J=7.7 Hz), 7.18 (1H, t, J=8.0 Hz), 6.66 (2H, brs), 6.46 (1H, s).
  • EXAMPLE 30 7-Amino-4-(2-trifluoromethylanilino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (300 mg, 1.56 mmol), 2-aminobenzotrifluoride hydrochloride (1.00 g, 5.06 mmol) and 2-aminobenzotrifluoride (2.00 g, 12.4 mmol) is stirred at 160° C. for 10 min. The resulting product is neutralized with excess NaHCO[0357] 3, dissolved in MeOH/CHCl3, dried onto silica gel and chromatographed over silica gel (6-7% MeOH/CH2Cl2) to give 7-amino-4-(2-trifluoromethylanilino)pyrido(4,3-d]pyrimidine (194 mg, 41%) as a cream solid, mp (MeOH/CHCl3/light petroleum) 126-130° C. (dec.). 1H NMR (DMSO) δ 10.60 (1H, brs), 9.17 (1H, brs), 8.13 (1H, brs), 7.76, 7.69 (1H, 1H, m, m), 7.45 (2H, m), 6.66 (2H, brs), 6.36 (1H, s).
  • EXAMPLE 31 7-Amino-4-(3-trifluoromethylanilino)pyrido(4,3-d]primidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (234 mg, 1.22 mmol) (described in a previous experimental) and 3-aminobenzotrifluoride (2.00 mL, 16.0 mmol) is stirred under N[0358] 2 at 190-200° C. for 2 h, and the resulting product is then chromatographed over silica gel (5-10% EtOH/EtOAc), and then over alumina (5-7% EtOH/CHCl3) to give 7-amino-4-(3-trifluoromethylanilino)pyrido[4,3-d]pyrimidine (157 mg, 42%) as a cream solid. 1H NMR (DMSO) δ 10.04 (1H, s), 9.37 (1H, s), 8.46 (1H, s), 8.31 (1H, s), 8.19 (1H, d, J=8.2 Hz), 7.62 (1H, t, J=8.0 Hz), 7.45 (1H, d, J=7.7 Hz), 6.69 (2H, brs), 6.47 (1H, s).
  • EXAMPLE 32 7-Amino-4-(4-trifluoromethylanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (390 mg, 2.03 mmol), 4-aminobenzotrifluoride hydrochloride (0.40 g, 2.02 mmol) and 4-aminobenzotrifluoride (1.61 g, 10.0 mmol) is stirred at 180° C. for 2 min. The resulting product is neutralized with excess NaHCO[0359] 3, dissolved in MeOH/CHCl3, dried onto alumina and chromatographed over alumina (4-7% MeOH/CH2Cl2) to give 7-amino-4-(4-trifluoromethylanilino)pyrido[4,3-d]pyrimidine (390 mg, 63%) as a cream solid. Analytically pure material was obtained by further chromatography over silica gel (5% MeOH/CH2Cl2) to give pale yellow needles. 1H NMR (DMSO) δ 10.09 (1H, brs), 9.40 (1H, s), 8.48 (1H, s), 8.13 (2H, d, J=8.2 Hz), 7.74 (2H, d, J=8.7 Hz), 6.72 (2H, brs), 6.40 (1H, s).
  • EXAMPLE 33 4- (3-Bromoanilino) -7-methylaminopyrido[4,3-d]pyrimidine
  • A mixture of 7-fluoro-4-(3-bromoanilino)pyrido[4,3d]pyrimidine (74 mg, 0.23 mmol), triethylamine (7 mL, 50 mmol) and methylamine hydrochloride (3.0 g, 44 mmol) in isopropanol (30 mL) contained in a steel bomb is stirred at 95° C. (oil bath) for 5 h. The resulting mixture is concentrated under vacuum, basified with aqueous Na[0360] 2CO3, diluted with water and extracted with EtOAc (3×100 mL). Chromatography of this extract on silica gel (3 a MeOH/CH2Cl2) gives 4-(3-bromoanilino)-7-methylaminopyrido[4,3-d]pyrimidine (50 mg, 65%) as a pale yellow solid. 1H NMR (DMSO) δ 9.93 (1H, brs), 9.37 (1H, s), 8.47 (1H, s), 8.18 (1H, s), 7.84 (1H, d, J=7.8 Hz), 7.34 (1H, t, J=7.9 Hz), 7.30 (1H, brd, J=8.1 Hz), 7.19 (1H, q, J=4.7 Hz), 6.35 (1H, s), 2.85 (3H, d, J=4.8 Hz).
  • EXAMPLE 34 4- (3-Bromoanilino) -7-dimethylaminoiyrido[4,3-d]pyrimidine
  • A mixture of 7-fluoro-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (101 mg, 0.32 mmol), triethylamine (4.4 mL, 32 mmole) and dimethylamine hydrochloride (2.58 g, 32 mmol) in isopropanol (30 mL) contained in a steel bomb is stirred at 100° C. (oil bath) for 4 h. The resulting solution is concentrated under vacuum, basified with aqueous Na[0361] 2CO3 and diluted with water to give a solid. Filtration and recrystallisation from MeOH/CHCl3 gives 7-dimethylamino-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (102 mg, 94%) as a pale yellow solid. 1H NMR (DMSO) δ 9.93 (1H, brs), 9.42 (1H, s), 8.48 (1H, s), 8.19 (1H, s), 7.85 (1H, d, J=7.7 Hz), 7.35 (1H, t, J=7.9 Hz), 7.30 (1H, brd, J=7.8 Hz), 6.53 (1H, s), 3.16 (6H, s).
  • EXAMPLE 35 4-[N-(3-Bromophenyl)-N-methylamino]-7-methylaminopyrido[4,3-d]pyrimidine
  • A mixture of 7-fluoro-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (100 mg, 0.31 mmole), triethylamine (4.4 mL, 32 mmole) and methylamine hydrochloride (2.12 g, 32 mmole) in isopropanol (30 mL) contained in a steel bomb is stirred at 100° C. (oil bath) for 5 h. The resulting mixture is concentrated under vacuum, basified with aqueous Na[0362] 2CO3, diluted with water and extracted with EtOAc (3×100 mL). Chromatography of this extract on silica gel (1-2% MeOH/CH2Cl2) gives 4-[N-(3-bromophenyl)-N-methylamino]-7-methylaminopyrido[4,3d]pyrimidine (23 mg, 21) as a pale yellow solid. 1H NMR (DMSO) δ 8.14 (1H, s), 7.79 (1H, s), 7.30 (1H, t, J=8.0 Hz), 7.20 (1H, ddd, J=7.9, 1.8, 0.8 Hz), 7.03 (1H, brq, J=4.9 Hz), 7.01 (1H, t, J=1.9 Hz), 6.82 (1H, ddd, J=7.8, 1.8, 0.9 Hz), 6.25 (1H, s), 3.40 (3H, s), 2.73 (3H, d, J=4.9 Hz).
  • EXAMPLE 36 7-Acetylamino-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (0.154 g, 0.49 mmol), acetic anhydride (0.14 mL, 1.5 mmol), triethylamine (0.14 mL, 1.0 mmol) and a catalytic amount of 4-(N,N-dimethylamino)pyridine are stirred under N[0363] 2 at room temperature for 18 h. The reaction is then quenched by addition of ice water. The dark precipitate is collected by Buchner filtration and is purified by preparative tlc (Rf=0.25, 7% MeOH/CHCl3). Recrystallization from EtOH gives 7-acetylamino-4-(3-bromoanilino)pyrido[4,3-d]-pyrimidine (13.5 mg, 7.7%). 1H NMR (DMSO) δ 10.92 (1H, s), 10.22 (1H, s), 9.64 (1H, s), 8.70 (1H, s), 8.28 (1H, s), 8.21 (1H,s), 7.88 (1H, d, J=7.7Hz) 7.41-7.34 (3H, m), 2.16 (3H, s).
  • EXAMPLE 37 4-(3-Bromoanilino)-7-methoxypyrido[4,3-d]pyrimidine
  • A solution of 7-fluoro-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (100 mg, 0.31 mmol) in 1 M sodium methoxide-methanol (30 mL) is stirred under reflux for 42 h. The resulting mixture is concentrated under reduced pressure, diluted with water and neutralized with dilute HCl to give 7-methoxy-4-(3-bromoanilino)pyrido[4,3-d]pyrimidine (92 mg, 89%) as a white solid. [0364] 1H NMR (DMSO) δ 10.22 (1H, brs), 9.57 (1H, s), 8.63 (1H, s), 8.19 (1H, s), 7.86 (1H, brd, J=7.9 Hz), 7.39 (1H, t, J=7.9 Hz), 7.35 (1H, dd, J=7.9, 1.5 Hz), 6.96 (1H, s), 4.00 (3H, s).
  • EXAMPLE 38 4-Benzylaminopyrido[4,3-d]pyrimidine
  • 4-Methylthiopyrido[4,3d]pyrimidine (160.4 mg, 0.902 mmol), and benzylamine (106.3 mg, 0.992 mmol) in EtOH (2 mL) are heated at 80° C. for 12 h, and then the solvent is removed under reduced pressure. The resulting solid is suspended in CH[0365] 2Cl2, filtered, and the resulting solid is purified by preparative tlc on silica, eluting with 5% MeOH in CHCl3. Removal of the solvent under reduced pressure yields 4-benzylaminopyrido[4,3-d]pyrimidine (36 mg, 17%). 1H NMR (DMSO) δ 9.60 (1H, s), 9.37 (1H, t, J=5.8 Hz), 8.72 (1H, d, J=5.8 Hz), 8.57 (1H, s), 7.54 (1H, d, J=5.8 Hz), 7.37 (2H, d, J=7.0 Hz), 7.33 (2H, t, J=7.3 Hz), 7.25 (1H, t, J=7.2 Hz), 4.81 (2H, d, J=5.8 Hz).
  • EXAMPLE 39 4-([R]-1-phenylethylamino)pyrido[4,3-d]pyrimidine
  • To a mixture of 4-methylthiopyrido[4,3-d]pyrimidine (85 mg, 0.48 mmol) and EtOH (2.5 mL) is added R-methylbenzylamine (0.13 mL, 1.0 mmol) dropwise. The resulting mixture is refluxed at 80° C. for 20 h. The solvent is removed under reduced pressure to give an oil which is crystallized from MeOH to give 4-([R]-1-phenylethylamino)pyrido[4,3-d]pyrimidine (41.6 mg, 35%), mp 138-138.5° C. [0366] 1H NMR (DMSO) δ 9.77 (1H, d, J=0.7 Hz), 9.00 (1H, d, J=7.7 Hz), 8.73 (1H, d, J=5.8 Hz), 8.54 (1H, s), 7.53 (1H, dd, J=5.8, 0.5 Hz), 7.45 (2H, d, J=7.2 Hz), 7.33 (2H, t, J=7.6 Hz), 7.23 (1H, tt, J 7.5, 1.2 Hz), 5.63 (1H, p, J=7.2 Hz), 1.61 (3H, t, J=7.0 Hz).
  • EXAMPLE 40 7-Amino-4-benzylaminopyrido[4,3-d]pyrimidine
  • A mixture of 2,4-diamino,5-cyanopyridinium acetate (8.78 g, 45 mmol), formic acid (10.66 g, 0.204 mol) and benzylamine (45 mL, 0.41 mol) is heated at 200° C. under N[0367] 2 for 2 h. Upon cooling, it solidifies. Water (500 mL) is added and the gummy solid/water mixture is stirred for −20 min. at 0° C. The liquid is decanted. The solid is washed with water and then recrystallized from isopropanol (25 mL). After drying in a vacuum oven overnight, 7-amino-2-benzylaminopyrido[4,3-d]pyrimidine (8.29 g, 73%) is obtained as a light yellow solid. 1H NMR (DMSO) δ 9.10 (1H, s), 8.85 (1H, t, J=5.8 Hz), 8.25 (1H, s), 7.21-7.36 (5H, m), 6.46 (2H, brs), 6.35 (1H, s), 4.74 (2H, d, J=6.0 Hz).
  • EXAMPLE 41 7-Amino-4-([R]-1-phenylethylamino)pyrido[4,3-d]pyrimidine
  • A mixture of [R]-1-phenylethylamine (0.072 mL, 0.55 mmol) and 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (97 mg, 0.5 mmol) (described in a previous experimental) is heated at 180° C. under N[0368] 2 for 1.5 hr. The reaction is then cooled to room temperature producing a precipitate. The mixture is added to water and CHCl3, sonicated and filtered. The phases are separated and the aqueous phase is extracted with CHCl3. The combined extracts are washed with water, saturated brine and dried (MgSO4). The solvent is removed under reduced pressure and the residue purified by using preparative tlc (5% MeOH/CHCl3) and recrystallization from CHCl3 to give 7-amino-4-([R]-1-phenylethylamino)pyrido[4,3-d]pyrimidine (14.5 mg, 11%), mp 231.8-232.1° C. 1H NMR (DMSO) δ 9.23(1H, s), 8.50 (1H, d, J=8.0 Hz), 8.19 (1H, s), 7.41 (2H, d, J=7.0 Hz), 7.31 (2H, t, J=8.0 Hz, 7.21 (1H, tt, J=7.4, 1.2 Hz), 6.45 (2H, s), 6.33 (1H, s), 5.56 (1H, p, J=7.2 Hz), 1.55 (3H, d, J=7.0 Hz).
  • EXAMPLE 42 7-Amino-4-(2-aminobenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (136 mg, 0.71 mmol) (described in a previous experimental) and 2-aminobenzylamine (1.70 g, 13.8 mmol) in isopropanol (5 mL) is stirred at reflux for 1 h, and the resulting product is chromatographed on silica gel (7-20% EtOH/EtOAc) and alumina (6-10% EtOH/CHCl[0369] 3) to give 7-amino-4-(2-aminobenzylamino)pyrido[4,3-d]pyrimidine (89 mg, 47%) as a white solid. 1H NMR (DMSO) δ 9.08 (1H, s), 8.68 (1H, t, J=5.8 Hz), 8.26 (1H, s), 7.05 (1H, d, J=7.4 Hz), 6.96 (1H, t, J=7.6 Hz), 6.63 (1H, d, J=7.9 Hz), 6.51 (1H, t, J=7.4 Hz), 6.46 (2H, brs), 6.35 (1H, s), 5.20 (2 H, brs), 4.56 (2H, d, J=5.8 Hz).
  • EXAMPLE 43 7-Amino-4-(3-dimethylaminobenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (236 mg, 1.23 mmol) (described in a previous experimental) and 3-dimethylamino-benzylamine (1.36 g, 9.07 mmol) in isopropanol (5 mL) is stirred under N[0370] 2 at reflux for 1 h, and the resulting product is chromatographed on silica gel (10-15% EtOH/EtOAc), then on alumina (1% EtOH/CHCl3) to give 7-amino-4-(3-dimethylaminobenzylamino)pyrido[4,3-d]pyrimidine (145 mg, 40%) as a white solid. 1H NMR (DMSO) δ 9.11 (1H, s), 8.79 (1H, t, J=5.9 Hz), 8.26 (1H, s), 7.11 (1H, dd, J=8.0, 7.7 Hz), 6.73 (1H, brs), 6.63 (1H, d, J=7.6 Hz), 6.60 (1H, dd, J=8.1, 2.2 Hz), 6.44 (2H, brs), 6.35 (1H, s), 4.67 (2H, d, J=5.8 Hz), 2.86 (6H, s).
  • EXAMPLE 44 7-Amino-4-(3-nitrobenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (228 mg, 1.19 mmol) (described in a previous experimental) and 3-nitrobenzylamine (0.81 g, 5.33 mmol) is stirred under N[0371] 2 at 150-160° C. for 1.5 h, and the resulting product chromatographed on silica gel (5-10% EtOH/EtOAc) to give 7-amino-4-(3-nitrobenzylmino)pyrido[4,3-d]pyrimidine (151 mg, 43%) as a yellow solid. 1H NMR (DMSO) δ 9.11 (1H, s), 8.98 (1H, t, J=5.5 Hz), 8.26 (1H, s), 8.22 (1H, brs), 8.12 (1H, dd, J=8.0, 1.8 Hz), 7.83 (1H, d, J=7.7 Hz), 7.63 (1H, t, J=7.9 Hz), 6.50 (2H, brs), 6.38 (1H, s), 4.85 (2H, d, J=5.8 Hz).
  • EXAMPLE 45 7-Amino-4-(3-methoxybenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (136 mg, 0.71 mmol) (described in a previous experimental) and 3-methoxybenzylamine (1.37 g, 10.0 mmol) in isopropanol (3 mL) is stirred under N[0372] 2 at reflux for 3 h. Evaporation of the solvent and chromatography on silica gel (5-10% EtOH/EtOAc) gives 7-amino-4-(3-methoxybenzylamino)pyrido[4,3-d]pyrimidine (153 mg, 77%) as a white solid. 1H NMR (DMSO) δ 9.11 (1H, s), 8.83 (1H, t, J=5.7 Hz), 8.26 (1H, s), 7.24 (1H, dt, Jd=0.8 Hz, J5=8.1 Hz), 6.92 (2H, m), 6.81 (1H, dt, Jd=8.2 Hz, Jt=1.2 Hz), 6.46 (2H, brs), 6.37 (1H, s), 4.71 (2H, d, J=5.8 Hz), 3.73 (3H, s).
  • EXAMPLE 46 7-Amino-4-(4-chlorobenzylamino)pyrido[4,3-d]pyrimidine mesylate
  • The free base (56 mg, 0.20 mmol)(prepared from 2,4-diamino,5-cyanopyridinium acetate, formic acid and 4-chlorobenzylamine at 200° C. as described in a previous example is precipitated from acetone solution with methanesulfonic acid (105 μL, 0.23 mmol) to give a polymesylate salt. [0373] 1H NMR (DMSO) δ 10.59 (1H, t, J=5.6 Hz), 9.24(1H, s), 8.69 (1H, s), 7.42 (4H, s), 6.42 (1H, s), 5.8 (6H, vbrs), 4.89 (2H, d, J=5.8 Hz), 2.41 (˜7.5H, s).
  • EXAMPLE 47 7-Amino-4-(2-bromobenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido(4,3-d]pyrimidine (225 mg, 1.17 mmol) (described in a previous experimental) and 2-bromobenzylamine (0.84 g, 4.52 mmol) is stirred under N[0374] 2 at 140° C. for 1 h, and the resulting product chromatographed on silica gel (1-5% EtOH/EtOAc) to give 7-amino-4-(2-bromobenzylamino)pyrido[4,3d]pyrimidine (175 mg, 45%) as a light brown solid. 1H NMR (DMSO) δ 9.16 (1H, s), 8.85 (1H, t, J=5.7 Hz), 8.24 (1H, s), 7.64 (1H, d, J=7.8 Hz), 7.34 (1H, dd, J=7.7, 7.1 Hz), 7.31 (1H, dd, J=7.7, 2.4 Hz), 7.21 (1H, ddd, J=7.8, 6.9, 2.4 Hz), 6.50 (2H, brs), 6.39 (1H, s), 4.74 (2H, d, J=5.7 Hz).
  • EXAMPLE 48 7-Amino-4-(3-bromobenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (228 mg, 1.19 mmol) (described in a previous experimental) and 3-bromobenzylamine (0.84 g, 4.52 mmol) is stirred under N[0375] 2 at 140° C. for 1 h. The resulting product is chromatographed on silica gel (2-10% EtOH/EtOAc) to give 7-amino-4-[(3-bromophenyl)methylamino]pyrido[4,3d]pyrimidine (203 mg, 52%) as a light brown solid. 1H NMR (DMSO) δ 9.09 (1H, s), 8.86 (1H, t, J=5.8 Hz), 8.26 (1H, s), 7.54 (1H, s), 7.44 (1H, d, J=7.8 Hz,), 7.36 (1H, d, J=7.6 Hz), 7.29 (1H, t, J=7.7 Hz), 6.48 (2H, s), 6.37 (1H, s), 4.73 (2H, d, J=5.8 Hz).
  • EXAMPLE 49 7-Amino-4-(4-bromobenzylamino)pyrido[4,3d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (234 mg, 1.22 mmol) (described in a previous experimental) and 4-bromobenzylamine (0.84 g, 4.52 mmol) is stirred under N[0376] 2 at 140° C. for 1 h, and the resulting product chromatographed on silica gel (10% EtOH/EtOAc) to give 7-amino-4-(4-bromobenzylamino)pyrido[4,3-d]pyrimidine (192 mg, 48%) as a cream solid. 1H NMR (DMSO) δ 9.09 (1H, s), 8.87 (1H, t, J=5.7 Hz), 8.25 (1H, s), 7.51 (2H, d, J=8.3 Hz), 7.31 (2H, d, J=8.3 Hz), 6.46 (2H, brs), 6.37 (1H, s), 4.70 (2H, d, J=5.8 Hz).
  • EXAMPLE 50 7-Amino-4-(2-trifluoromethylbenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (225 mg, 1.17 mmol) and 2-(trifluoromethyl)benzylamine (0.90 mL, 6.42 mmol) is stirred under N[0377] 2 at 150° C. for 1 h. The resulting product is chromatographed on silica gel (5% EtOH/EtOAc) to give 7-amino-4-(2-trifluoromethylbenzyl)aminopyrido[4,3d]pyrimidine (0.22 g, 59%) as a white solid. 1H NMR (DMSO) δ 9.16 (1H, s), 8.88 (1H, t, J=5.7 Hz), 8.23 (1H, s), 7.75 (1H, d, J=7.7 Hz), 7.62 (1H, t, J=7.5 Hz), 7.50 (1H, d, J=7.4 Hz), 7.47 (1H, t, J=7.6 Hz), 6.51 (2H, brs), 6.39 (1H, s), 4.92 (2H, d, J=5.5 Hz).
  • EXAMPLE 51 7-Amino-4-(3-trifluoromethylbenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (225 mg, 1.17 mmole) and 3-(trifluoromethyl)benzylamine (0.63 mL, 4.40 mmole) is stirred under N[0378] 2 at 140° C. for 1 h. The resulting product is chromatographed on silica gel (3-5% EtOH/EtOAc) to give 7-amino-4-[(3-trifluoromethylphenyl)methylamino]pyrido[4,3-d]pyrimidine (0.24 g, 63%) as a light brown solid. 1H NMR (DMSO) δ 9.10 (1H, s), 8.92 (1H, t, J=5.7 Hz), 8.26 (1H, s), 7.71 (1H, s), 7.66 (1H, d, J=7.4 Hz), 7.62 (1H, d, J=7.8 Hz), 7.57 (1H, t, J=7.6 Hz), 6.49 (2H, brs), 6.38 (1H, s), 4.82 (2H, d, J=5.8 Hz).
  • EXAMPLE 52 7-Amino-4-(4-trifluoromethylbenzylamino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (225 mg, 1.17 mmol) and 4-(trifluoromethyl)benzylamine (0.63 mL, 4.42 mmol) is stirred under N[0379] 2 at 140° C. for 1 h. The resulting product is chromatographed on alumina (5-10% EtOH/CHCl3) then silica gel (2-10% EtOH/EtOAc) to give 7-amino-4-[(4-trifluoromethylphenyl)methylamino]pyrido[4,3-d]pyrimidine (0.21 g, 56%) as a light brown solid. 1H NMR (DMSO) δ 9.12 (1H, s), 8.94 (1H, t, J=5.8 Hz), 8.24 (1H, s), 7.69 (2H, d, J=8.1 Hz), 7.56 (2H, d, J=8.1 Hz), 6.48 (2H, brs), 6.38 (1H, s), 4.82 (2H, d, J=5.8 Hz).
  • EXAMPLE 53 7-Amino-4-(thien-2-ylmethylamino)[4,3-d]pyrimidine dimesylate
  • The compound is obtained from 2,4-diamino,5-cyanopyridinium acetate (190 mg, 0.98 mmol), formic acid (0.23 g, 4.4 mmol) and thienylmethylamine (1.07 ml, 10 mmol) as described in a previous experimental. The crude product is converted into a dimesylate salt as described previously and recrystallized from Pr[0380] iOH to give 7-amino-4-(thien-2-ylmethylamino)pyrido[4,3-d]pyrimidine dimesylate in 19% yield. 1H NMR (DMSO δ 10.67 (1H, t, J=5.8 Hz), 9.21 (1H, s), 8.77 (1H, s), 7.48 (1H, dd, J=5.1, 1.2 Hz) 7.16 (1H, dd, J=3.4, 0.7 Hz), 7.02 (1H, dd, J=4.8, 3.4 Hz), 6.42 (1H, s), 5.06 (2H, d, J=5.7 Hz), 2.41 (6H, s).
  • EXAMPLE 54 7-Acetylamino-4-benzylaminopyrido[4,3-d]pyrimidine
  • 7-Acetylamino-4-methylthiopyrido[4,3-d]pyrimidine. Acetyl chloride (0.70 mL, 9.84 mmol) is added to a solution of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (0.20 g, 1.04 mmol) (described in a previous experimental) and Et[0381] 3N (1.51 mL, 10.8 mmol) in THF at 0° C., and then the mixture is stirred at 20° C. for 4 h. Water (50 mL) was added, then the solution was extracted with EtOAc (3×50 mL). Evaporation and chromatography on alumina (18 EtOH/CHCl3) yields 7-acetylamino-4-methylthiopyrido[4,3-d]pyrimidine (0.12 g, 49%) as a yellow solid,. 1H NMR (DMSO) δ 11.05 ( 1H, s), 9.30 (1H, s), 9.02 (1H, s), 8.38 (1H, s), 2.71 (3H, s) , 2.18 (3H, s)
  • 7-Acetylamino-4-benzylaminopyrido[4,3-d]pyrimidine. A mixture of 7-acetylamino-4-methylthiopyrido[4,3-d]pyrimidine (0.40 g, 1.71 mmol) and benzylamine (1.0 mL, 9.15 mmol) is stirred under N[0382] 2 at 140° C. for 1 h, and the resulting product is chromatographed on silica gel (EtOAc) to give 7-acetylamino-4-benzylaminopyrido[4,3-d]pyrimidine (0.31 g, 62%) as a white solid. 1H NMR (DMSO) δ 10.79 (1H, s), 9.42 (1H, s), 9.23 (1H, t, J=5.8 Hz), 8.49 (1H, s), 8.18 (1H, s), 7.39 (1H, dt, Jd=6.9 Hz, Jt=1.7 Hz), 7.34 (1H, tt, J=7.3, 1.7 Hz), 7.25 (1H, tt, J=7.1, 1.7 Hz), 4.80 (2H, d, J=5.8 Hz), 2.15 (3H, s).
  • EXAMPLE 55 4-Anilinopyrido[3,4-d]pyrimidine
  • 4-Carboxamidonicotinic acid. 3,4-Pyridine dicarboxylic anhydride (8.3 g, 55.6 mmol) is added to conc NH[0383] 4OH (12 mL) in H2O (60 mL) stirred at 0° C. over 5 min. Upon addition a paste forms which is stirred for 1 h at room temperature. The white paste is sparged with N2 for 30 min and diluted with H2O (10 mL) to form a clear solution. Then SO2 is bubbled through the solution for 15 min reducing its pH to 2. Upon cooling the resulting solid is filtered, rinsed with H2O, and oven dried to yield 4-carboxamidonicotinic acid (7 g, 76%) as a white solid. 1H NMR (DMSO) δ 8.93 (1H, s), 8.76 (1H, d, J=5.0 Hz), 8.08 (1H, s), 7.62 (1H, s), 7.45 (1H, d, J=5.0 Hz).
  • Isocuinolinic imide. 4-Carboxamidonicotinic acid (280 mg, 1.68 mmol) is heated neat at 200° C. for 5 h to yield isoquinolinic imide (177.2 mg, 71%) as a white solid. [0384] 1H NMR (DMSO) δ 11.68 (H, s)), 9.12-9.03 (2H, m), 7.80 (1H, d, J=5.1 Hz).
  • 3-Amino isonicotinic acid. Bromine (1.71 g) is added to 10% KOH (30 mL) on ice. The resulting solution is added to finely ground isoquinolinic imide (1.46 g, 9.86 mmol). Upon addition the mixture begins to foam. When all of the solid is dissolved up aqueous KOH (15%, 7 mL) is added and the mixture is heated to 80° C. for 1 min then cooled. The mixture is neutralized with SO[0385] 2, and cooled to 0° C. until precipitation occurs. The solid is collected by suction filtration and washed with H2O, and dried in a vacuum oven to yield of 3-amino isonicotinic acid (485 mg, 36%) as a white solid. 1H NMR (DMSO) δ 9.5-8.8 (2H, brs), 8.20 (1H, s), 7.70 (1H, d, J=5 Hz), 7.46 (1H, d, J=5 Hz)
  • 3H-Pyrido[3,4-d]pyrimid-4-one. A mixture of 3-amino isonicotinic acid (485 mg, 3.51 mmol) in formamide (3 mL) is heated to 160° C. for 12 h. Upon cooling, the resulting solid is filtered and washed with H[0386] 2O and dried in a vacuum oven to yield 3H-pyrido[3,4-d]pyrimid-4-one (373 mg, 72%). 1H NMR (DMSO) δ 12.60 (1H, brs), 9.06 (1H, s), 8.68 (1H, d, J=5.3 Hz), 8.23 (1H, s), 7.96 (1H, d, J=5.1 Hz).
  • 4-Thiopyrido[3,4-d]pyrimidine. Phosphorous pentasulfide (1.25 g, 2.74 mmol) is added to a solution of 3H-pyrido[3,4-d]pyrimid-4-one (366 mg, 2.49 mmol) in pyridine (4 mL). The mixture is refluxed for 4 h under N[0387] 2. The resulting black tar is dissolved in H2O, and a solid forms. The solid is filtered and washed with H2O and dried in a vacuum oven to yield 4-thiopyrido[3,4-d]pyrimidine (369.8 mg, 91%) as a yellow solid. 1H NMR (DMSO) δ 14.48 (1H, brs), 9.13 (1H,s), 8.70 (1H, d, J=5.4 Hz), 8.29 (1H, s), 8.27 (1H, d, J=5.4 Hz).
  • 4-Methylthiopyrido[3,4-d]pyrimidine. A mixture of 4-thiopyrido[3,4d]pyrimidine (369.8 mg, 2.26 mmol), triethylamine (0.6 mL, 4.5 mmol), DMSO (2 mL), and iodomethane (0.24 mL, 3.96 mmol) is stirred under N2 at 25° C. for 12 h. The mixture is poured into H[0388] 2O and the resulting solid is filtered and dried in a vacuum oven to yield 4-methylthiopyrido[3,4-d]pyrimidine (222 mg, 55) as a brown solid. 1H NMR (DMSO) δ 9.51 (1H, s) , 9.18 (1H, s), 8.79 (1H, d, J=8 Hz), 7.97 (1H, d, J=8 Hz).
  • 4-Anilinopyrido[3,4-d]pyrimidine A mixture of 4-methylthiopyrido[3,4d]pyrimidine (75 mg, 0.42 mmol), and aniline (1 mL) is heated to 100° C. under N[0389] 2 for 2 h. The reaction mixture is then chromatographed on silica using MPLC and eluting with a gradient system (CHCl3 to 5% MeOH in CHCl3). The fractions are concentrated under reduced pressure, and the resulting solid is recrystallized from Et2O to yield 4-anilinopyrido[3,4-d]pyrimidine (21.2 mg, 23%) as a yellow solid. 1H NMR (DMSO) δ 10.09 (1H, s), 9.18 (1H, s), 8.74 (1H, d, J=5.3 Hz), 8.46 (1H, d, J=5.8 Hz), 7.89 (2H, d, J=8.5 Hz), 7.45 (2H, t, J=7.9 Hz), 7.21 (1H, t, J=7.4 Hz).
  • EXAMPLE 56 4-(3-Bromoanilino)pyrido[3,4-d]pyrimidine
  • A mixture of 4-methylthiopyrido[3,4-d]pyrimidine (75 mg, 0.42 mmol) (see previous experimental), and 3-bromoaniline (1 mL) is heated to 100° C. under N[0390] 2 for 2 h. The reaction mixture is then chromatographed on silica using MPLC and eluting with a gradient system (CHCl3 to 5% MeOH in CHCl3). The fractions are concentrated under reduced pressure, and the resulting solid is recrystallized from Et2O to yield 4-(3-bromoanilino)pyrido[3,4d]pyrimidine (66 mg, 52.7%) as a light brown solid. 1H NMR (DMSO) δ 10.15 (1H, s), 9.21 (1H, s), 8.80 (1H, s), 8.76 (1H, d, J=5.8 Hz), 8.44 (1H, d, J=5.6 Hz), 8.25 (1H, s), 7.93 (1H, d, J=7.7 Hz), 7.45-7.37 (2H, m).
  • EXAMPLE 57 4-(3-Bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine
  • 5-[N-(tert-Butoxycarbonyl)amino]-2-fluoropyridine. 5-Amino-2-fluoropyridine is prepared by hydrogenation (Pd/C) of 2-fluoro-5-nitropyridine (obtained from from 2-chloro-5-nitropyridine by reaction with KF in MeCN with Ph[0391] 4PBr [J. H. Clark and D. J. Macquarrie, Tetrahedron Lett., 1987, 28, 111-114]. Reaction of the crude amine with t-Boc anhydride gives 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine. 1H NMR (CDCl3 δ 8.07 (1H, s), 8.05 (1H, m), 6.89 (1H, dd, J=9.2, 3.3 Hz), 6.66 (1H, m), 1.52 (9 H, s).
  • 5-[N-(tert-Butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid. Reaction of 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine (5.3 g, 25 mmol) sequentially with n-BuLi and CO[0392] 2 as described in the following example gives 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid (1.60 g, 25%). 1H NMR (DMSO) δ 9.83 (1H, brs), 8.84 (1H, s), 7.49 (1H, d, J=2.9 Hz), 1.47 (9H, s).
  • 5-Amino-2-fluoropyridine-4-carboxylic acid. Reaction of 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid (1.0 g, 3.9 mmol) with TFA as described above gives 5-amino-2-fluoropyridine-4-carboxylic acid(0.46 g, 74%). [0393] 1H NMR (DMSO) δ 7.85 (1H, d, J=1.5 Hz), 7.23 (1H, d, J=2.5 Hz).
  • 6-Fluoro-3H-pyrido[3,4-d]pyrimidin-4-one. Reaction of 5-amino-2-fluoropyridine-4-carboxylic acid with formamide at 140° C. as above gave 6-fluoro-3H-pyrido[3,4-d]pyrimidin-4-one (−20%). [0394] 1H NMR (DMSO) δ 12.48 (1H, m), 8.74 (1H, s), 8.16 (1H, s), 7.63 (1H, d, J=3 Hz).
  • 4-(3-Bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine. Reaction of 6-fluoro-3H-pyrido[3,4-d]pyrimidin-4-one (0.60 g, 3.6 mmol) with POCl[0395] 3, followed by reaction of the crude 4,6-dihalo compound with 3-bromoaniline gives 4-(3-bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine (0.73 g, 63%). 1H NMR (DMSO) δ 10.09 (1H, brs), 8.96 (1H, s), 8.75 (1H, s), 8.25 (2H, m), 7.90 (1H, brd, J=6.5 Hz), 7.44-7.34 (2H, m).
  • EXAMPLE 58 4-(3-Bromoanilino)-6-chloropyrido[3,4-d]pyrimidine
  • 5-[N-(tert-butoxycarbonyl)amino]-2-chloropyridine. A mixture of 5-amino-2-chloropyridine (12.86 g, 0.1 mol), di-tert-butyldicarbonate (24.0 g, 0.11 mol) and Et[0396] 3N (12.1 g, 1.12 mol) in CH2Cl2 (150 mL) is heated under reflux for 12 h, cooled, and the precipitate is filtered off. The organic layer is washed with water, dried (CaCl2) and filtered through a short column of alumina. Removal of the solvent gives 5-[N-(tert-butoxycarbonyl)amino]-2-chloropyridine (11.9 g, 52%). 1H NMR (CDCl3) δ 8.31 (1H, d, J=2.9 Hz), 7.94 (1H, dd, J=8.6, 2.6 Hz), 7.24 (1H, d, J=8.7 Hz), 7.15 (1H, m), 1.51 (9 H, s).
  • 5-[N-(tert-Butoxycarbonyl)amino]-2-chloroyridine-4-carboxylic acid. A solution of 5-[N-(tert-butoxycarbonyl)amino]-2-chloropyridine (22.87 g, 0.1 mol) and TMEDA (47 mL, 0.31 mol) in dry Et[0397] 2O (600 mL) is cooled to −78° C., and n-BuLi (10 M in hexanes, 30 mL, 0.3 mol) is added dropwise. The solution is allowed to warm to −10° C. and is then kept at that temperature for 2 h, before being recooled to −78° C. Dry CO2 is then bubbled in, and the resulting mixture is allowed to warm to 20° C., before being quenched with water (300 mL) containing a small amount of NH4OH. The resulting aqueous layer is washed with EtOAc, then acidified slowly with dilute HCl to precipitate 5-[N-(tert-butoxycarbonyl)amino]-2-chloropyridine-4-carboxylic acid (15.5 g, 57%). 1H NMR (DMSO) δ 10.00 (H, s), 9.13 (1H, s), 7.74 (1H, s), 1.47 (9H, s).
  • 5-Amino-2-chloropyridine-4-carboxylic acid. A stirred suspension of 5-[N-(tert-butoxycarbonyl)amino]-2-chloropyridine-4-carboxylic acid (1.91 g, 7 mmol) in CH[0398] 2Cl2 (200 mL) is treated slowly with trifluoroacetic acid until homogeneous (ca. 12 mL). The solution is stirred overnight and extracted with dilute NH4OH, and the aqueous layer is then acidified with dilute HCl to gave a precipitate of 5-amino-2-chloropyridine-4-carboxylic acid (1.05 g, 87% yield). 1H NMR (DMSO) δ 9.01 (2H, m) , 8.03 (1H, s) 7.48 (1H, s).
  • 6-Chloro-3H-pyrido[3,4-d]pyrimidin-4-one. A solution of 5-amino-2-chloropyridine-4-carboxylic acid (8.1 g, 4.7 mmol) in formamide (100 mL) is stirred at 140° C. for 12 h. Dilution of the cooled mixture with water gives a precipitate of 6-chloro-3H-pyrido[3,4-d]pyrimidin-4-one (7.3 g, 86% yield). [0399] 1H NMR (DMSO) δ 12.73 (1H, m), 8.90 (1H, d, J=0.7 Hz), 8.23 (1H, s. 7.97 (1H, d, J=0.7 Hz).
  • 4,6-Dichloropyrido[3,4-d]pyrimidine. A stirred suspension of 6-chloropyrido[3,4-d]pyrimidin-4-one (1.82 g, 10 mmol) in POCl[0400] 3 (10 mL) is heated under reflux until dissolved (ca. 2 h) and for a further 30 min. Excess reagent is removed under reduced pressure, and the residue is treated with a mixture of CH2Cl2 and ice-cold aqueous Na2CO3. The resulting organic layer is dried (Na2SO4) and evaporated to give a quantitative yield of crude, unstable, 4,6-dichloropyrido[3,4-d]pyrimidine, which is used directly in the next step. 1H NMR (CDCl3) δ 9.38 (1H, d, J=0.5 Hz), 9.19 (1H, s), 8.09 (1H, d, J=0.5 Hz).
  • 4-(3-Bromoanilino)-6-chloropyrido[3,4-d]pyrimidine. A mixture of the above crude dichloropyrimidine and 3-bromoaniline (3.8 g, 22 mmol) is dissolved in i-PrOH (100 mL). One drop of conc. HCl is added to initiate the reaction, and the mixture is then heated under reflux for 30 min, cooled, and diluted with water to precipitate 4-(3-bromoanilino)-6-chloropyrido[3,4-d]pyrimidine (1.26 g, 38% yield). 1H NMR (DMSO) δ 10.12 (1H, s), 9.03 (1H, s), 8.77 (1H, s), 8.63 (1H, s), 8.21 (1H, s), 7.89 (1H, d, J=8.1 Hz) , 7.43-7.32 (2H, m). [0401]
  • EXAMPLE 59 4-(3-Bromoanilino)-6-methoxypyrido[3,4-d]pyrimidine
  • Treatment of 4-(3-bromoanilino)-6-fluoropyrido[3,4d]pyrimidine (see a previous experimental) at 100° C. in a pressure vessel with sodium methoxide in methanol gives 4-(3-bromoanilino)-6-methoxypyrido[3,4-d]pyrimidine. [0402] 1H NMR (DMSO) δ 9.93 (1H, s), 8.94 (1H, s), 8.61 (1H, s), 8.26 (1H, brs), 7.94 (1H, brd, J=7.6 Hz), 7.88 (1H, s), 7.43-7.32 (2H, m), 4.01 (3H, s).
  • EXAMPLE 60 4-(3-Bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine
  • Treatment of 4-(3-bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine (0.20 g, 0.63 mmol)(see a previous experimental) at 100° C. in a pressure vessel with methylamine in ethanol followed by chromatography on alumina (CH[0403] 2Cl2/MeOH, 99:1) gives 4-(3-bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine (0.07 g, 34%). 1H NMR (DMSO) δ 9.69 (1H, s), 8.75 (1H, s), 8.41 (1H, s), 8.21 (1H, brs), 7.93 (1H, brd, J=7.6 Hz), 7.41-7.28 (2H, m), 7.06 (1H, s), 6.82 (1H, q, J=5.0 Hz), 4.95 (3H, d, J=5.0 Hz).
  • EXAMPLE 61 4-(3-Bromoanilino)-6-dimethylaminopyrido[3,4-d]pyrimidine
  • Treatment of 4-(3-bromoanilino)-6-fluoropyrido[3,4d]pyrimidine (see a previous experimental) at 100° C. in a pressure vessel with dimethylamine in ethanol gives 4-(3-bromoanilino)-6-dimethylaminopyrido[3,4-d]pyrimidine. [0404] 1H NMR (DMSO) δ 9.71 (1H, s), 8.83 (1H, s), 8.43 (1H, s), 8.21 (1H, brs), 7.94 (1H, brd, J=7.5 Hz), 7.42-7.29 (2H, m), 7.26 (1H, s), 3.17 (6H, s).
  • EXAMPLE 62 4-(Benzylamino)pyrido[3,4-d]pyrimidine
  • A mixture of 4-methylthiopyrido[3,4-d]pyrimidine (74 mg, 0.41 mmol)(see a previous experimental), and benzylamine (1 mL) is heated to 100° C. for 2 h. On cooling the mixture is concentrated under reduced pressure and purified directly by preparative tlc on silica gel eluting with CH[0405] 2Cl2, to yield 4-(benzylamino)pyrido[3,4-d]pyrimidine (21.2 mg, 20%). 1H NMR (DMSO) δ 9.21 (1H, t, J=5.8 Hz), 9.19 (1H, s), 8.63 (1H, d, J=5.8 Hz), 8.58 (1H, s), 8.20 (1H, d, J=5.1 Hz), 7.41-7.30 (4H, m), 7.26 (1H, t, J=7.1 Hz).
  • EXAMPLE 63 4-(3-Bromoanilino)pyrido[2,3-d]pyrimidine
  • 3H-pyrido[2,3-d]pyrimidin-4-one. 2-Amino nicotinic acid (15 g, 108.6 mmol) in formamide (35 mL) is heated to 165-170° C. for 3.5 h. Upon cooling a solid precipitates. The solid is filtered and washed with H[0406] 2O and dried in a vacuum oven to give 3H-pyrido[2,3-d]pyrimidin-4-one (7.87 g, 49.4%). 1H NMR (DMSO) δ 12.50 (1H, s), 8.97 (1H, dd, J=1.9, 4.5 Hz), 8.53 (1H, dd, J=2.1, 7.9 Hz), 8.34 (1H, s), 7.57 (1H, dd, J=4.6, 8.0 Hz).
  • 4-Thiopyrido[2,3-d]pyrimidine. Phosphorous pentasulfide (6 g, 13.5 mmol) is added to a solution of 3H-pyrido[2,3-d]pyrimidin-4-one (2 g, 13.5 mmol) in pyridine (50 mL). The mixture is refluxed for 3 h. Upon cooling a solid formed and the pyridine is decanted off. The solid is suspended in H[0407] 2O (20 mL) and then filtered and dried in a vacuum oven to yield 4-thiopyrido[2,3-d]pyrimidine (1.72 g, 78%). 1H NMR (DMSO)δ 9.06 (1H, dd, J=1.9, 4.3 Hz), 8.90 (1H, dd, J=1.9, 8.2 Hz), 8.36 (1 H, s), 7.65 (1H, dd, J=4.3, 8.2 Hz).
  • 4-Methylthiopyrido[2,3-d]pyrimidine. A mixture of 4-thiopyrido[2,3-d]pyrimidine (100 mg, 0.76 mmol), triethylamine (154 mg, 1.52 mmol), DMSO (2 mL), and iodomethane (161 mg, 1.14 mmol) is stirred for 12 h at 25° C. The mixture is poured into H[0408] 2O and extracted with EtOAc. The combined extracts are washed with water, saturated brine, and dried (MgSO4), and the solvent is removed under reduced pressure to yield 4-methylthiopyrido[2,3d]pyrimidine (134 mg, quant.). 1H NMR (DMSO) δ 9.25 (1H, dd, J=1.8, 4.2 Hz), 9.17 (1H, s), 8.59 (1H, dd, J=1.9, 8.2 Hz), 7.75 (1H, dd, J=4.3, 8.2 Hz), 2.73 (3H, s).
  • A mixture of 4-methylthiopyrido[2,3-d]pyrimidine (157 mg, 0.89 mmol, and 3-bromoaniline (1 mL) is heated to 100° C. for 2 h. On cooling a precipitate forms which is filtered then washed with EtOH and air dried to yield 4-(3-bromoanilino)pyrido[2,3-d]pyrimidine (55.5 mg, 20%. [0409] 1H NMR (DMSO) δ 10.13 (1H, s), 9.11 (1H, dd, J=1.7, 4.3 Hz), 9.01 (1H, dd, J=1.7, 8.2 Hz), 8.81 (1H, s), 8.22 (1H, s), 7.90 (1H, d, J=7.7 Hz), 7.71 (1H, dd, J=4.3, 8.0 Hz), 7.40 (2H, m)
  • EXAMPLE 64 4-(3-Bromoanilino)-7-fluoropyrido[2,3-d]pyrimidine
  • 2,6-Difluoronicotinic acid. 2,6-Difluoropyridine (7.89 mL, 0.087 mmol) is added dropwise under N[0410] 2 at 78° C. to a stirred solution of lithium diisopropylamide (59.0 mL of a 1.5 N solution in cyclohexane, 0.089 mmol) in THF (250 mL). After 2 h at 78° C., a stream of dry CO2 is passed through the solution and the mixture is diluted with water and washed with EtOAc. The aqueous portion is neutralized with 3 N HCl, extracted with EtOAc and worked up to give 2,6-difluoronicotinic acid (13.4 g, 97%) . 1H NMR (DMSO) δ 8.59 (1H, dd, J=9.2, 8.2 Hz), 7.30 (1H, dd, J=8.2, 2.1 Hz), 4.03 (1H, brs).
  • 2,6-Difluoronicotinamide. A [0411] solution 2,6-difluoronicotinic acid (7.4 g, 0.046 mmol) and SOCl2 (20 mL) in 1,2-dichloroethane (60 mL) containing DMF (1 drop) is heated under reflux for 4 h, then concentrated to dryness under reduced pressure. The residue is dissolved in Et2O (100 mL), cooled to 0° C., and treated dropwise with concentrated ammonia (10.0 mL, 0.17 mmol). After 10 min the solution is washed with aqueous NaHCO3 and worked up to give 2,6-difluoronicotinamide (5.61 g, 76%). 1H NMR (CDCl3) δ 8.70 (1H, dd, J=9.6, 8.3 Hz), 7.00 (1H, ddd, J=8.3, 2.9, 1.1 Hz), 6.71, 6.55 (1H, 1H, 2 brs).
  • 2-Amino-6-fluoronicotinamide. A solution of 2,6-difluoronicotinamide (4.68 g, 0.029 mmol) in dry formamide (30 mL) is saturated with ammonia and allowed to stand at room temperature for 24 h. Water (50 mL) is added and the resultant precipitate is filtered off and washed well with water, to give 6-amino-2-fluoronicotinamide (1.41 g, 31%) mp 236-237° C. [0412] 1H NMR (DMSO) δ 7.89 (1H, dd, J=10.4, 8.4 Hz), 7.31, 7.16 (1H, 1H, 2 brs,), 6.93 (2H, brs), 6.36 (1H, dd, J=8.4, 2.4 Hz).
  • The filtrate and washings are combined and extracted exhaustively with EtOAc, and the extract is chromatographed on silica gel. EtOAc/petroleum ether (1:1) elutes forerun, while EtOAc/petroleum ether (2:1) and then EtOAc gives 2-amino-6-fluoronicotinamide (1.57 g, 35%), mp (EtOAc/petroleum ether) 199-200° C. (Rogers, R. B. et al., U.S. Pat. No. 4,383,851, record mp 198-200° C.]. [0413] 1H NMR (DMSO) δ 8.13 (1H, dd, J=10.4, 8.4 Hz), 7.90, 7.30 (1H, 1H, 2 brs), 7.65 (2H, brs), 6.23 (1H, dd, J=8.4, 2.6 Hz).
  • A suspension of 2-amino-6-fluoronicotinamide (0.74 g, 4.77 mmol) in triethyl orthoformate (25 mL) is heated at reflux for 8 h. After cooling to room temperature the precipitate is filtered off and washed well with petroleum ether to give 7-fluoropyrido[2,3-d]pyrimid-4(3H)-one (0.76 g, 96%),. [0414] 1H NMR (DMSO) δ 12.75 (1H, brs), 8.66 (1H, dd, J=10.4, 8.4 Hz), 8.38 (1H, s), 7.33 (1H, dd, J=8.4, 2.6 Hz).
  • 4-(3-Bromoanilino)-7-fluoropyrido[2,3-d]pyrimidine. A suspension of 7-fluoropyrido[2,3-d]pyrimid-4(3H)-one (0.20 g, 1.21 mmol) in POCl[0415] 3 (10 mL) is heated under reflux for 2 h. The volatiles are then removed under reduced pressure, and the residue is partitioned between aqueous NaHCO3 and EtOAc. The organic extract is worked up to give crude 4-chloro-7-fluoropyrido[2,3-d]pyrimidine, which is used directly in the next reaction. A solution of this product (0.20 g, 1.09 mmol) and 3-bromoaniline (0.23 mL, 2.18 mmol) in propan-2-ol (1.0 mL) and THF (10 mL) containing a trace of conc. HCl is stirred at 20° C. for 1 h, and then concentrated to dryness. The residue is dissolved in EtOAc, washed with aqueous NaHCO3, and worked up to give an oil, which is chromatographed on silica gel. Elution with EtOAc/petroleum ether (1:5) gives 3-bromoaniline, while EtoAc/petroleum ether (1:1) elutes 4-(3-bromoanilino)-7-fluoropyrido[2,3-d]pyrimidine (0.18 g, 47%), mp (MeOH) 211-213° C. 1H NMR (DMSO) δ 10.18 (1H, brs), 9.17 (1H, t, J=8.6 Hz), 8.80 (1H, s), 8.17 (1H, t, J=1.8 Hz), 7.85 (1H, dt, Jd=7.6 Hz, Jt=1.8 Hz), 7.53 (1H, dd, J=8.6, 2.7 Hz), 7.41-7.34 (2H, m).
  • EXAMPLE 65 7-Amino-4-(3-bromoanilino)pyrido[2,3-d]pyrimidine
  • A solution of 4-(3-bromoanilino)-7-fluoropyrido[2,3d]pyrimidine (0.20 g, 0.63 mmol) in EtOH (20 mL) is saturated with ammonia and warmed at 100° C. in a pressure vessel for 30 h. The solvent is removed under reduced pressure to give 7-amino-4-(3-bromoanilino)pyrido[2,3-d]pyrimidine (0.18 g, 90%). [0416] 1H NMR (DMSO) δ 9.97 (1H, brs), 8.59 (1H, s), 8.51 (1H, d, J=9.3 Hz), 8.11 (1H, sl brs), 7.77 (1H, brd, J=6.3 Hz), 7.44 (2H, brs), 7.37-7.30 (2H, m), 6.81 (1H, d, J=9.3 Hz).
  • EXAMPLE 66 4-(3-Bromoanilino)-7-methylaminopyrido[2,3-d]pyrimidine
  • A solution of 4-(3-bromoanilino)-7-fluoropyrido[2,3d]pyrimidine (see a previous experimental) (0.20 g, 0.63 mmol), methylamine hydrochloride (0.13 g, 1.88 mmol) and Et[0417] 3N (0.30 mL) 2.19 mmol) in EtOH (15 mL) is heated at 100° C. in a pressure vessel for 18 h. The solvent is removed under reduced pressure, and the residue is partitioned between EtOAc and water. Workup of the organic layer gives 4-(3-bromoanilino)-7-(methylamino)pyrido[2,3-d]pyrimidine (0.16 g, 77%). 1H NMR (DMSO) δ 9.53 (1H, s), 8.54 (1H, s), 8.41 (1H, d, J=8.1 Hz), 8.17 (1H, t, J=1.8 Hz), 7.83 (1H, dd, J=8.0, 1.9 Hz), 7.66 (1H, brs), 7.32 (1H, t, J=8.0 Hz), 7.24 (1H, dd, J=8.0, 1.8 Hz), 6.77 (1H, d, J=8.1 Hz), 2.92 (3H, d, J=4.8 Hz).
  • EXAMPLE 67 4-(3-Bromoanilino)-7-dimethylaminopyrido[2,3-d]pyrimidine
  • Reaction of 4-(3-bromoanilino)-7-fluoropyrido[2,3-d]pyrimidine (see a previous experimental) (0.12 g, 0.38 mmol) with dimethylamine hydrochloride (92 mg, 1.13 mmol) and Et[0418] 3N (0.18 mL, 1.32 mmol) in EtOH (15 mL) at 100° C. for 18 h in a pressure vessel, followed by evaporation of the solvent and workup, gives 4-(3-bromoanilino)-7-(dimethylamino)pyrido[2,3-d]pyrimidine (0.11 g, 84%). 1H NMR (DMSO) δ 9.58 (1H, brs), 8.56 (1H, d, J=9.3 Hz), 8.54 (1H, s), 8.18 (1H, t, J=1.9 Hz), 7.84 (dt, Jd=8.0, Hz, J5=1.9 Hz), 7.33 (1H, dd, J=8.1, 8.0 Hz) 7.25 (1H, dt, Jd=9-3, Hz, Jt=1.9 Hz), 7.10 (1H, d, J=9.3 Hz), 3.18 (6H, s).
  • EXAMPLE 68 4-(3-Bromoanilino)-7-methoxypyrido[2,3-d]pyrimidine
  • A solution of 4-(3-bromoanilino)-7-fluoropyrido[2,3-d]pyrimidine (0.26 g, 0.81 mmol) and sodium methoxide (prepared from 75 mg of sodium, 3.26 mmol) in dry MeOH (15 mL) is heated at 90° C. in a pressure vessel for 18 h. The mixture is poured into water and extracted with EtOAc to give 4-(3-bromoanilino)-7-methoxypyrido[2,3d]pyrimidine (0.23 g, 86%). [0419] 1H NMR (DMSO) δ 9.88 (1H, brs), 8.82 (1H, d, J=8.9 Hz), 8.71 (1H, s), 8.18 (1H, dd, J=8.0, 1.9 Hz), 7.36 (1H, dd, J=8.1, 8.0 Hz), 7.29 (1H, ddd, J=8.1, 1.9, 1.9 Hz) 7.15 (1H, d, J=8.9 Hz), 4.01 (3H, s).
  • EXAMPLE 69 4-Benzylamino-7-methylaminopyrimido[4,5-d]pyrimidine
  • S-Ethylisothiouronium iodide. A solution of thiourea (3.80 g, 50 mmol) and iodoethane (4 mL, 50 mmol) in MeOH (100 mL) is refluxed for 24 h. The solvent is stripped under reduced pressure, and the residual light yellow oil, is dried under vacuum, solidifying spontaneously. The desired compound (13.98 g) is obtained quantitatively. [0420]
  • 4-Amino-5-cyano-2-ethylthiopyrimidine A suspension of NaOMe (2.7 g, 50 mmol) in EtOH (200 mL) is added to a mixture of S-ethylisothiourea hydroiodide (11.58 g, 50 mmol), ethoxymethylidenemalononitrile (6.1 g, 50 mmol) and ethanol (250 mL) at 25° C. The reaction mixture is refluxed under N[0421] 2 for 2 h, and then the solution is concentrated on a hot plate until precipitation is observed. After cooling, the solid is collected by suction filtration and is stirred in water at 25° C. Filtration and vacuum oven drying affords 4-amino-5-cyano-2-ethylthiopyrimidine (4.02 g, 45a) as a brown solid. 1H NMR δ (DMSO) 8.45 (1H, s), 7.90 (2H, brs), 3.00 (2H, q, J=7.3 Hz), 1.27 (3H, t, J=7.3 Hz).
  • 4-Amino-2-ethylthiopyrimidine-5-carboxamide. 4-Amino-5-cyano-2-ethylthiopyrimidine (4.0 g, 22.3 mmol) is added to sulfuric acid (conc., 4.3 mL) in small portions. The mixture is then stirred under N[0422] 2 at 40° C. for 1.5 h. The reaction is quenched with ice-water and NH4OH is used to adjust the pH to ˜9. The solid is collected via suction filtration and dried in a vacuum oven overnight. 4-Amino-2-ethylthiopyrimidine-5-carboxamide (2.58 g, 58%) is obtained as a light brown solid. 1H NMR: (DMSO) δ 8.52 (1H, s), 7.98 (2H, brs), 7.42 (2H, brs), 3.04 (2H, q, J=7.3 Hz), 1.27 (3H, t, J=7.3 Hz).
  • 4-Oxo-7-ethylthio-3H-pyrimido[4,5-d]pyrimidine. A mixture of 4-amino-2-ethylthiopyrimidine-5-carboxamide (4.66 g, 23.5 mmol) and triethyl orthoformate (150 mL) is refluxed under N[0423] 2 for 24 h, and is then cooled to 25° C. The brown solid is isolated by suction filtration and dried in a vacuum oven to give 4-oxo-7-ethylthio-3H-pyrimido[4,5-d]pyrimidine (3.54 g, 72). 1H NMR: (DMSO) δ 12.80 (1H, s), 9.20 (1H, s), 8.45 (1H, s), 3.18 (2H, q, J=7.4 Hz), 1.35 (3H, t, J=7.4 Hz).
  • 4-Thiono-7-ethylthio-3H-pyrimido[4,5-d]pyrimidine. A mixture of 4-oxo-7-ethylthio-3H -pyrimido[4,5-d]pyrimidine (1.33 g, 6.7 mmol), P[0424] 2S5 (1.48 g, 6.6 mmol) and pyridine (15 mL) is refluxed under N2 for 3 h. The pyridine is then stripped under reduced pressure, and the residue is dissolved in NaOH solution (0.5 M, 75 mL) and boiled with charcoal. After filtration, the filtrate is neutralized with acetic acid to generate a gold brown solid. Buchner filtration and drying in a vacuum oven affords 4-thiono-7-ethylthio-3H-pyrimido[4,5d]pyrimidine (1.42 g, 95%). 1H NMR (DMSO) δ 9.47 (1H, s), 8.46 (1H, s), 3.20 (2H, q, J=7.3 Hz), 1.35 (3H, t, J=7.3 Hz).
  • 7-Ethylthio-4-methylthiopyrimido[4,5-d]pyrimidine. The same procedure described for 7-amino-4-methylthiopyrido[4,3d]pyrimidine in Example 21 is used. [0425] 1H NMR (DMSO) δ 9.52(1H, s), 9.15 (1H, s), 3.23 (2H, q, J=7.3 Hz), 2.72 (3H, s), 1.38 (3H, t, J=7.3 Hz).
  • 4-Benzylamino-7-ethylthiopyrimido[4,5-d]pyrimidine. The same procedure described for 7-amino-4-anilinopyrido[4,3-d]pyrimidine in example 21 is used. [0426]
  • 4-Benzylamino-7-methylaminopyrimido[4,5-d]pyrimidine. 4-Benzylamino-7-ethylthiopyrimido[4,5-d]pyrimidine in EtOH containing excess methylamine is heated to 150° C. in a stainless steel bomb for 5 h. The solid is filtered off and dried to give 4-benzylamino-7-methylaminopyrimido[4,5-d]pyrimidine. [0427]
  • EXAMPLE 70 4-Benzylamino-7-hydrazinopyrimido[4,5-d]pyrimidine
  • 4-Benzylamino-7-ethylthio pyrimido[4,5-d]pyrimidine in EtOH containing excess hydrazine is heated to 150° C. in a stainless steel bomb for 5 h. The solid is filtered off and dried to give 4-benzylamino-7-hydrazinopyrimido[4,5-d]pyrimidine. [0428]
    Figure US20010027197A1-20011004-C00015
  • EXAMPLE 71 4-(3-Bromoanilino)thieno[3,2-d]pyrimidine hydrochloride
  • 3H-Thieno[3,2-d]pyrimid-4-one. A mixture of methyl 3-aminothiophene-2-carboxylate (1 g, 6.3 mmol) and formamide (2 g) is heated at 240° C. for 10 min. Upon cooling a precipitate appeared. It is dissolved in EtOH and filtered. The filtrate is concentrated under reduced pressure and the residue is purified by silica gel chromatography eluting with 10% MeOH in CH[0429] 2Cl2 to yield 3H-thieno[3,2-d]pyrimid-4-one (249 mg, 26%) as a solid. 1H NMR (DMSO) δ 12.61 (1H, brs), 8.20 (1H, s), 8.17 (1H, d, J=5 Hz), 7.42 (1H, d, J=5 Hz).
  • 4-Chlorothieno[3,2-d]pyrimidine. To a solution of DMF (170.3 μL, 2.2 mmol) and dichloroethane (1.2 mL) at 0° C. under N[0430] 2, oxalyl chloride (279.2 mg, 3.2 mmol) is added slowly and stirred for 10 min. 3H-thieno[3,2-d]pyrimid-4-one (152.2 mg, 1.0 mmol) is added and refluxed for 5 h. The reaction mixture is poured into water and extracted with CH2Cl2. The organic layer is stripped under reduced pressure to yield 4-chlorothieno[3,2-d]pyrimidine (140 mg, 82%) as a yellow solid. 1H NMR (DMSO) δ 9.05 (1H, s), 8.62 (1H, d, J=5 Hz), 7.79 (1H, d, J=5 Hz).
  • 4-(3-Bromoanilino)thieno[3,2-d]pyrimidine hydrochloride. A mixture of 4-chlorothieno[3,2-d]-pyrimidine (135 mg, 0.79 mmol) and 3-bromoaniline (95 μL, 0.89 mmol) in 2-methoxyethanol (2 mL) is heated to 79° C. for 30 min. The resulting precipitate is filtered and washed with CH[0431] 2Cl2 to yield 4-(3-bromoanilino)thieno[3,2-d]pyrimidine hydrochloride (195.5 mg, 72%) as a light yellow solid. 1H NMR (DMSO) δ 11.33 (1H, s), 8.94 (1H, s), 8.23 (1H, s), 8.53 (1H, d, J=5.3 Hz), 8.07 (1H, s), 7.77 (1H, d, J=7.9 Hz), 7.6 (1H, d, J=5.3 Hz), 7.48 (2H, m).
  • EXAMPLE 72 4-Benzylaminothieno[3,2-d]pyrimidine
  • As described in the previous experiment 4-chlorothieno[3,2-d]pyrimidine (100 mg, 0.586 mmol) and benzylamine (710 μL, 0.645 mmol) in 2-methoxyethanol (2 mL) yields 4-benzylaminothieno[3,2-d]pyrimidine (37 mg, 26%). [0432] 1H NMR (DMSO) δ 8.42 (1H, s), 8.12 (1H, d, J=5.5 Hz), 7.39 (1H, d, J=5.3 Hz), 7.40-7.30 (4H, m), 7.24 (1H, t, J=6.8 Hz).
  • EXAMPLE 73 4-(3-Bromoanilino)thieno[2,3-d]pyrimidine
  • Methyl 2-aminothiophene-3-carboxylate. A mixture of methyl cyanoacetate (3.25 g, 32.3 mmol), 1,4 dithiane-2,5 diol (5 g, 32.8 mmol), triethylamine (1 mL, 7.71 mmol) in EtOH (50 mL) is stirred at 40° C. for 1 h. The cooled solution is eluted through a silica plug with CH[0433] 2Cl2 . The filtrate is stripped to dryness to give crude methyl 2-aminothiophene-3-carboxylate which is carried on to the next reaction. 1H NMR (DMSO) δ 7.26 (1H, s), 6.82 (1H, d, J=5.8 Hz), 6.28 (1H, d, J=5.8 Hz), 3.69 (3H, s).
  • 3H-Thieno[2,3-d]pyrimid-4-one. A solution of methyl 2-aminothiophene-3-carboxylate (602.1 mg, 3.83 mmol) in formamide (5 mL) is heated at 200° C. for 12 h. The resulting tar is dissolved in CH[0434] 2Cl2 (10 mL) then placed on a silica plug and eluted with 10% MeOH in CH2Cl2. The filtrate is stripped under reduce pressure and the resulting solid is washed with EtOH to yield 3H-thieno[2,3-d]pyrimid-4-one (231.4 mg, 40%) as an orange solid. 1H NMR (DMSO) δ 12.50 (1H, brs), 8.13 (1H, s), 7.60 (1H, d, J=5.8 Hz), 7.41 (1H, d, J=6.0 Hz).
  • 4-Chlorothieno[2,3-d]pyrimidine. To a solution of DMF (90 μL) and CH[0435] 2Cl2 (2 mL) at 0° C. under N2, oxalyl chloride (148 mg, 1.2 mmol) is added slowly and stirred for 10 min. 3H-Thieno[2,3-d]pyrimid-4-one (81 mg, 0.52 mmol) is added as a solid to the solution and warmed with a heat gun until the solid dissolves. The reaction is stirred at 25° C. for 12 h under N2. The reaction mixture is poured into water and extracted with CH2Cl2 . The phases are separated and the organic layer is dried (Na2SO4) and stripped under reduced pressure to yield 4-chlorothieno[2,3-d]pyrimidine (87.6 mg, 97%) as a solid. 1H NMR (DMSO) δ 8.96 (1H, s), 8.17 (1H, d, J=6.0 Hz), 7.62 (1H, d, J=6.0 Hz).
  • 4-(3-Bromoanilino)thieno[2,3d]pyrimidine hydrochloride. A mixture of 4-chlorothieno[2,3-d]pyrimidine (135 mg, 0.79 mmol) and 3-bromoaniline (95 μL, 0.89 mmol) in 2-methoxyethanol (2 mL) is heated to 79° C. for 30 min with stirring. The resulting solid is filtered and washed with CH[0436] 2Cl2 to yield 4-(3-bromoanilino)thieno[2,3d]pyrimidine hydrochloride (197 mg, 73%). 1H NMR (DMSO) δ 9.99 (1H, s), 8.60 (1H, s), 8.23 (1H, s), 7.98 (1H, d, J=6.0 Hz), 7.88 (1H, d, J=8.0 Hz), 7.79 (1H, d, J=6.0 Hz), 7.37 (1H, t, J=8.0 Hz), 7.30 (1H, d, J=8.0 Hz).
  • EXAMPLE 74 4-Benzylaminonyrrolo[2,3d]pyrimidine
  • 4-Benzylaminopyrrolo[2,3-d]pyrimidine is prepared as described previously. G. H. Hitchings, K. W. Ledig and R. A. West, U.S. Pat. No. 3,037,980, 1962; Chemical Abstracts 1962, 57, 15130c. [0437]
  • EXAMPLE 75 N6-(3-Bromophenyl)adenine
  • A mixture of 6-chloropurine (1.0 g, 6.47 mmol), 3-bromoaniline (0.78 mL, 7.12 mmol), and conc HCl (4 drops) in isopropanol (10 mL) is stirred at 80° C. for 5 h. Upon cooling, it precipitates. The solid is filtered and washed with isopropanol and air dried to yield N[0438] 6-(3-bromophenyl)adenine (1.93 g, 91%) as a light yellow solid. 1H NMR (DMSO) δ 11.38 (1H, s), 8.78 (1H, s), 8.75 (1H, s), 7.90 (1H, d, J=8.0 Hz), 7.38-7.34 (2H, m).
    Figure US20010027197A1-20011004-C00016
  • EXAMPLE 76 N6 -Benzyladenine
  • N[0439] 6-Benzyladenine is available commercially from the Aldrich Chemical Company, 1001 West Saint Paul Avenue, Milwaukee, Wis. 53233.
  • EXAMPLE 77 7-Amino-4-(3-methylanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido [4,3-d]pyrimidine (217 mg, 1.13 mmol) and m-toluidine (1.50 g, 14.0 mmol) is stirred at 155° C. for 30 min. The resulting product is chromatographed over silica gel (5% MeOH/CH[0440] 2Cl2) to give 7-amino-4-(3-methylanilino)pyrido[4,3d]pyrimidine (190 mg, 67%) as a pale yellow solid. 1H NMR (DMSO) δ 9.81 (1H, brs), 9.34 (1H, s), 8.38 (1H, s), 7.60 (2H, s), 7.26 (1H, dd, J=8.5, 7.6. Hz), 6.95 (1H, d, J=7.4 Hz), 6.63 (2H, brs), 6.44 (1H, s), 2.33 (3H, s).
  • EXAMPLE 78 7-Amino-4-(4-methoxyanilino)pyrido[4,3-d]pyrimidine
  • A mixture of 7-amino-4-methylthiopyrido[4,3-d]pyrimidine (129 mg, 0.62 mmol) and 4-methoxyaniline ((0.15 g, 1.2 mmol) was in ethanol (5 mL) was heated at 40° C. for 16 h, and then reflux for 3 h. The reaction mixture was cooled to 0° C. overnight, and the solid was colected by vacuum filtration and recrystalized from isopropanol to give 7-amino-4-(4-methoxyanilino)pyrido[4,3-d]pyrimidine (42 mg, 25%) as a yellow solid. [0441] 1H NMR (DMSO) δ 10.00 (1H, brs), 9.31 (1H, s), 8.35 (1H, s), 7.62 (2H, d, J=9.2 Hz), 6.96 (2H, d, J=9.2 Hz), 6.70 (2H, slbrs), 6.41 (1H, s), 3.77 (3H, s).
  • EXAMPLE 79 4-(3-Bromoanilino)-6-(piperidin-1-yl)pyrido[3,4-d]pyrimidine
  • Treatment of 4-(3-bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine (see a previous experimental) at 100° C. in a pressure vessel with piperidine in ethanol gives 4-(3-bromoanilino)-6-dimethylaminopyrido[3,4-d]pyrimidine. [0442]
  • The pharmaceutical compositions of the invention can take any of a wide variety of oral and parenteral dosage forms. The dosage forms comprise as the active components an inhibitor as defined previously. [0443]
  • For preparing pharmaceutical compositions, one uses inert, pharmaceutically acceptable carriers that can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets, and suppositories. A solid carrier can be one or more substances which may also act as dilutents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material. In powders, the carrier is a finely divided solid which is in admixture with the finely divided active compounds. In the tablet, the active compounds are mixed with carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired. The powders and tablets preferably contain from 5% or 10% to about 70% of active ingredients. Suitable solid carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low melting wax, cocoa butter, and the like. The term “preparation” is intended to include the formulation of the active compounds with encapsulating materials as carrier, providing a capsule in which the active components (with or without other carriers) are surrounded by carrier, which are thus in association with it. Similarly, cachets are included. Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration. [0444]
  • Liquid form preparations include solutions, suspensions, and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection. Liquid preparations can also be formulated in solution in aqueous polyethylene glycol solution. Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizing, and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active components in water with viscous material, i.e., natural or synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other well-known suspending agents. [0445]
  • Preferably, the pharmaceutical preparation is in unit dosage form. In such form, the preparation may be subdivided into unit doses containing appropriate quantities of inhibitor and other anti-cancer materials individually or as a combination, i.e., in a mixture. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, for example, packeted tablets, capsules, and powders in vials or ampoules. The unit dosage form can also be a capsule, cachet, or tablet itself or it can be the appropriate number of any of these in packaged form. Additionally, the unit dosage form may be a dividable form having an inhibitor in one part and other anti-cancer materials in the other part, such as, a dividable capsule, a dividable package, or a two-part ampoule, vial or the like. [0446]
  • The quantity of an inhibitor in unit dosages of preparation may be varied or adjusted from about 0.01 mg/kg to 100.0 mg/kg, preferably 0.03 mg/kg to less than 1.0 mg/kg of inhibitor. [0447]
  • The pharmaceutical compositions preferably are constituted so that they can be administered parenterally or orally. Solutions of the active compounds as free bases and free acids or pharmaceutically acceptable salts can be prepared in water suitable mixed with a surfactant such as hydroxypropylcellulosa. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. [0448]
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of the microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, paragens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferred to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions of agents delaying absorption, for example, gelatin. [0449]
  • Sterile-injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients, into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of the sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze-drying technique which yields a powder of active ingredients plus an additional desired ingredient from a previously sterile-filtered solution thereof. [0450]
  • As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. [0451]
  • It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suitable as unitary dosages for the mammalian subjects to be treated,; each unit containing a predetermined quantity of active materials calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active materials and the particular therapeutic effect to be achieved, and (b) the limitation inherent in the art of compounding such active materials for the treatment of disease in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail. [0452]
  • The principal active ingredients are compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in dosage unit form as hereinbefore disclosed. A unit parenteral dosage form can, for example, contain the principal active compound, i.e. an inhibitor, in amounts ranging from about 0.5 to about 100 mg, with from about 0.1 to 50 mg being preferred. The daily parenteral doses for mammalian subjects to be treated ranges from 0.01 mg/kg to 10 mg/kg of the inhibitor. The preferred daily dosage range is 0.1 mg/kg to 1.0 mg/kg. [0453]
  • For oral dosages, the daily amount may range from 0.01 mg of active compound/kg of mammalian subject to 100 mg/kg, preferably 0.1 to 10 mg/kg of subject. [0454]
  • The inhibitor described above may form commonly known, pharmaceutically acceptable salts such as alkali metal and other common basic salts or acid addition salts, etc. References to the base substances are therefore intended to include those common salts known to be substantially equivalent to the parent compound and hydrates thereof. [0455]
  • The active compounds described herein are capable of further forming both pharmaceutically acceptable acid addition and/or base salts. All of these forms are within the scope of the present invention. [0456]
  • Pharmaceutically acceptable acid addition salts of the active compounds include salts derived from nontoxic inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, hydrofluoric, phosphorous, and the like, as well as the salts derived from nontoxic organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. Such salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, trifluoroacetate. propionate, caprylate, isobutyrate, oxalate, malonate succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like. Also contemplated are salts of amino acids such as arginate and the like and gluconate, galacturonate (see, for example, Berge, S. M. et al, “Pharmaceutical Salts”, [0457] JOURNAL OF PHARMACEUTICAL SCIENCE, 66, pp. 1-19 (1977)).
  • The acid addition salts of said basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner. Preferably, an active compound can be converted to an acidic salt by treating with an aqueous solution of the desired acid, such that the resulting pH is less than 4. The solution can be passed through a C18 cartridge to absorb the compound, washed with copious amounts of water, the compound eluted with a polar organic solvent such as, for example, methanol, acetonitrile, and the like, and isolated by concentrating under reduced pressure followed by lyophilization. The free base form may be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner. The free base forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base for purposes of the present invention. [0458]
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge, S. M. et al, “Pharmaceutical Salts”, [0459] JOURNAL OF PHARMACEUTICAL SCIENCE, 66, pp. 1-19 (1977)).
  • The base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. Preferably, an active compound can be converted to a base salt by treating with an aqueous solution of the desired base, such that the resulting pH is greater than 9. The solution can be passed through a C18 cartridge to absorb the compound, washed with copious amounts of water, the compound eluted with a polar organic solvent such as, for example, methanol, acetonitrile and the like, and isolated by concentrating under reduced pressure followed by lyophilization. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner. The free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acids for purposes of the present invention. [0460]
  • Certain of the compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms, including hydrated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. [0461]
  • Certain of the compounds of the present invention possess one or more chiral centers and such center may exist in the R(D) or S(L) configuration. The present invention includes all enantiomeric and epimeric forms as well as the appropriate mixtures thereof. [0462]
  • While the forms of the invention herein constitute presently preferred embodiments, many others are possible. It is not intended herein to mention all of the possible equivalent forms or ramifications of the invention. It is understood that the terms used herein are merely descriptive rather than limiting and that various changes may be made without departing from the spirit or scope of the invention. [0463]
    Figure US20010027197A1-20011004-C00017
    Figure US20010027197A1-20011004-C00018
    Figure US20010027197A1-20011004-C00019
    Figure US20010027197A1-20011004-C00020
    Figure US20010027197A1-20011004-C00021
    Figure US20010027197A1-20011004-C00022
    Figure US20010027197A1-20011004-C00023
    Figure US20010027197A1-20011004-C00024
    Figure US20010027197A1-20011004-C00025
    Figure US20010027197A1-20011004-C00026
    Figure US20010027197A1-20011004-C00027
    Figure US20010027197A1-20011004-C00028
    Figure US20010027197A1-20011004-C00029
    Figure US20010027197A1-20011004-C00030
    Figure US20010027197A1-20011004-C00031
    Figure US20010027197A1-20011004-C00032
    Figure US20010027197A1-20011004-C00033
    Figure US20010027197A1-20011004-C00034
    Figure US20010027197A1-20011004-C00035
    Figure US20010027197A1-20011004-C00036
    Figure US20010027197A1-20011004-C00037
    Figure US20010027197A1-20011004-C00038
    Figure US20010027197A1-20011004-C00039
    Figure US20010027197A1-20011004-C00040
    Figure US20010027197A1-20011004-C00041
    Figure US20010027197A1-20011004-C00042
    Figure US20010027197A1-20011004-C00043
    Figure US20010027197A1-20011004-C00044
    Figure US20010027197A1-20011004-C00045
    Figure US20010027197A1-20011004-C00046
    Figure US20010027197A1-20011004-C00047
    Figure US20010027197A1-20011004-C00048
    Figure US20010027197A1-20011004-C00049

Claims (34)

What is claimed is:
1. A method of inhibiting epidermal growth factor receptor tyrosine kinase by treating, with an effective inhibiting amount, a mammal, in need thereof, a compound of Formula II:
Figure US20010027197A1-20011004-C00050
where:
one of A or E is nitrogen, with remaining atoms carbon;
X=O, S, NH or NR7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms);
n=0, 1, 2;
R1=H or lower alkyl (1-4 carbon atoms); if n=2, R1 can be independently H or lower alkyl (1-4 carbon atoms) on either linking carbon atom;
R2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcycloalkyl (3-8 carbon atoms), sulfonylcycloalkyl (3-8 carbon atoms), sulfonamido, lower mono or dialkylsulfonamido (1-4 carbon atoms), mono or dicycloalkylsulfonamido (3-8 carbon atoms), mercapto, carboxy, carboxamido (—C(O)—NH2), lower mono or dialkylcarboxamido (1-4 carbon atoms), mono or dicycloalkylcarboxamido (3-8 carbon atoms), lower alkoxycarbonyl (1-4 carbon atoms), cycloalkoxycarbonyl (3-8 carbon atoms), lower alkenyl (2-4 carbon atoms), cycloalkenyl (4-8 carbon atoms), lower alkynyl (2-4 carbon atoms), or two R2 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring; and
m=0-3,
Ar is selected from the group consisting of phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl;
R3, R4, R5 and R6 are independently selected from the group consisting of H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms); carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms);
lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, and lower O-alkylhydroxylamino (1-4 carbon atoms);
or optionally R3 and R4 or R4 and R6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
any lower alkyl group substituent on any of the substituents in R3-R6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
R8 is hydrogen, lower alkyl (1-4 carbon atoms), amino or mono or dialkyl (1-4 carbon atoms) amino;
with the proviso that R3 cannot be either OH or SH;
with the further proviso that at least one of the R3 and R4 or R4 and R6 substituents must be other than hydrogen, halogen, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms);
optionally if any of the substituents R1, R2, R3, R4 or R6 have chiral centers, then all stereoisomers thereof both as racemic and/or diastereoisomeric mixtures are included;
or a pharmaceutical salt or hydrate thereof.
2. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower alkoxy or halogen.
3. The method of
claim 1
wherein X=NH, n =0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one amino.
4. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
5. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 and R4 lower alkyl.
6. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
7. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
8. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen and R3 lower mono or dialkylamino, with R4 hydroxy.
9. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, B, D & E carbon, with A nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
10. The method of
claim 1
having any one of the following ring structures:
Figure US20010027197A1-20011004-C00051
11. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower alkoxy.
12. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one amino.
13. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower mono or dialkylamino.
14. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one hydrazino.
15. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 or R4 H, with the other one lower alkyl.
16. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkoxy.
17. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R3 and R4 lower alkyl.
18. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 amino, with the other one lower alkoxy.
19. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
20. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R4 lower mono or dialkylamino, with R3 hydroxy.
21. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R2 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
22. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 or R4 lower mono or dialkylamino, with the other one lower alkoxy.
23. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen and R4 lower mono or dialkylamino, with R3 hydroxy.
24. The method of
claim 1
wherein X=NH, n=0 or 1, in which case R1=H, the aromatic ring phenyl optionally substituted, A, B & D carbon, with E nitrogen, and R3 and R4 taken together are dioxymethylene, dioxyethylene, 2,3-fused piperazine, 2,3-fused morpholine or 2,3-fused thiomorpholine.
25. A compound of Formula II
Figure US20010027197A1-20011004-C00052
where:
one of A or E is nitrogen, with remaining atoms carbon;
X=O, S, NH or NR7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms);
n=0, 1, 2;
R1=H or lower alkyl (1-4 carbon atoms); if n=2, R1 can be independently H or lower alkyl (1-4 carbon atoms) on either linking carbon atom;
R2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcycloalkyl (3-8 carbon atoms), sulfonylcycloalkyl (3-8 carbon atoms), sulfonamido, lower mono or dialkylsulfonamido (1-4 carbon atoms), mono or dicycloalkylsulfonamido (3-8 carbon atoms), mercapto, carboxy, carboxamido (—C(O)—NH2), lower mono or dialkylcarboxamido (1-4 carbon atoms), mono or dicycloalkylcarboxamido (3-8 carbon atoms), lower alkoxycarbonyl (1-4 carbon atoms), cycloalkoxycarbonyl (3-8 carbon atoms), lower alkenyl (2-4 carbon atoms), cycloalkenyl (4-8 carbon atoms), lower alkynyl (2-4 carbon atoms), or two R2 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring; and
m=0-3,
Ar is selected from the group consisting of phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl;
R3, R4, R5 and R6 are independently selected from the group consisting of H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms); carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms);
lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, and lower O-alkylhydroxylamino (1-4 carbon atoms);
or optionally R3 and R4 or R4 and R6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
any lower alkyl group substituent on any of the substituents in R3-R6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
R8 is hydrogen, lower alkyl (1-4 carbon atoms), amino or mono or dialkyl (1-4 carbon atoms) amino;
with the proviso that R3 cannot be either OH or SH;
with the further proviso that at least one of the R3 and R4 or R4 and R6 substituents must be other than hydrogen, halogen, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms);
with the further proviso that Formula II excludes ring structures where A or B is nitrogen when the A-E ring is a ring having 6 members;
optionally if any of the substituents R1, R2, R3, R4 or R6 have chiral centers, then all stereoisomers thereof both as racemic and/or diastereoisomeric mixtures are included;
or a pharmaceutical salt or hydrate thereof.
26. A pharmaceutical composition adapted for administration as an inhibitor of the epidermal growth factor receptor family of tyrosine kinases, comprising a therapeutically effective amount of a compound of Formula I or Formula II in admixture with a pharmaceutically acceptable excipient, diluent or carrier:
Figure US20010027197A1-20011004-C00053
where:
one of A or E is nitrogen, with remaining atoms carbon;
X=O, S, NH or NR7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms);
n=0, 1, 2;
R1=H or lower alkyl (1-4 carbon atoms); if n=2, R1 can be independently H or lower alkyl (1-4 carbon atoms) on either linking carbon atom;
R2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcycloalkyl (3-8 carbon atoms), sulfonylcycloalkyl (3-8 carbon atoms), sulfonamido, lower mono or dialkylsulfonamido (1-4 carbon atoms), mono or dicycloalkylsulfonamido (3-8 carbon atoms), mercapto, carboxy, carboxamido (—C(O)—NH2), lower mono or dialkylcarboxamido (1-4 carbon atoms), mono or dicycloalkylcarboxamido (3-8 carbon atoms), lower alkoxycarbonyl (1-4 carbon atoms), cycloalkoxycarbonyl (3-8 carbon atoms), lower alkenyl (2-4 carbon atoms), cycloalkenyl (4-8 carbon atoms), lower alkynyl (2-4 carbon atoms), or two R2 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring; and
m=0-3,
Ar is selected from the group consisting of phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl;
R3, R4, R5 and R6 are independently selected from the group consisting of H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms); carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms);
lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, and lower O-alkylhydroxylamino (1-4 carbon atoms);
or optionally R3 and R4 or R4 and R6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
any lower alkyl group substituent on any of the substituents in R3-R6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
R8 is hydrogen, lower alkyl (1-4 carbon atoms), amino or mono or dialkyl (1-4 carbon atoms) amino;
with the proviso that R3 cannot be either OH or SH;
with the further proviso that at least one of the R3 and R4 or R4 and R6 substituents must be other than hydrogen, halogen, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms);
with the further proviso that Formula II excludes ring structures where A or B is nitrogen when the A-E ring is a ring having 6 members;
optionally if any of the substituents R1, R2, R3, R4 or R6 have chiral centers, then all stereoisomers thereof both as racemic and/or diastereoisomeric mixtures are included;
or a pharmaceutical salt or hydrate thereof.
27. A method of treating cancer by treating, with an effective cancer inhibiting amount, a mammal, in need thereof, a compound of Formula I or Formula II:
Figure US20010027197A1-20011004-C00054
where:
one of A or E is nitrogen, with remaining atoms carbon;
X=O, S, NH or NR7, such that R7=lower alkyl (1-4 carbon atoms), OH, NH2, lower alkoxy (1-4 carbon atoms) or lower monoalkylamino (1-4 carbon atoms);
n=0, 1, 2;
R1=H or lower alkyl (1-4 carbon atoms); if n=2, R1 can be independently H or lower alkyl (1-4 carbon atoms) on either linking carbon atom;
R2 is lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), nitro, halo (fluoro, chloro, bromo, iodo), lower perfluoroalkyl (1-4 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms; —O—C(O)R), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms), hydroxymethyl, lower acyl (1-4 carbon atoms; —C(O)R), cyano, lower thioalkyl (1-4 carbon atoms), lower sulfinylalkyl (1-4 carbon atoms), lower sulfonylalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), sulfinylcycloalkyl (3-8 carbon atoms), sulfonylcycloalkyl (3-8 carbon atoms), sulfonamido, lower mono or dialkylsulfonamido (1-4 carbon atoms), mono or dicycloalkylsulfonamido (3-8 carbon atoms), mercapto, carboxy, carboxamido (—C(O)—NH2), lower mono or dialkylcarboxamido (1-4 carbon atoms), mono or dicycloalkylcarboxamido (3-8 carbon atoms), lower alkoxycarbonyl (1-4 carbon atoms), cycloalkoxycarbonyl (3-8 carbon atoms), lower alkenyl (2-4 carbon atoms), cycloalkenyl (4-8 carbon atoms), lower alkynyl (2-4 carbon atoms), or two R2 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring; and
m=0-3,
Ar is selected from the group consisting of phenyl, thienyl, furanyl, pyrrolyl, pyridyl, pyrimidyl, imidazoyl, pyrazinyl, oxazolyl, thiazolyl, naphthyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl and quinazolinyl;
R3, R4, R5 and R6 are independently selected from the group consisting of H, lower alkyl (1-4 carbon atoms), cycloalkyl (3-8 carbon atoms), lower alkoxy (1-4 carbon atoms), cycloalkoxy (3-8 carbon atoms), hydroxy, lower acyloxy (1-4 carbon atoms), amino, lower mono or dialkylamino (1-4 carbon atoms), lower mono or dicycloalkylamino (3-8 carbon atoms); carbonato (—OC(O)OR) where the R is lower alkyl of 1 to 4 carbon atoms or cycloalkyl of 3-8 carbon atoms;
ureido or thioureido or N- or O-linked urethane any one of which is optionally substituted by mono or di-lower alkyl (1-4 carbon atoms) or cycloalkyl (3-8 carbon atoms);
lower thioalkyl (1-4 carbon atoms), thiocycloalkyl (3-8 carbon atoms), mercapto, lower alkenyl (2-4 carbon atoms), hydrazino,N′-lower alkylhydrazino (1-4 carbon atoms), lower acylamino (1-4 carbon atoms), hydroxylamino, and lower O-alkylhydroxylamino (1-4 carbon atoms);
or optionally R3 and R4 or R4 and R6 taken together on contiguous carbon atoms can form a carbocyclic ring of 5-7 members or a monounsaturated 1,3-dioxolanyl, 1,4-dioxanyl, 1,4-dioxepinyl, pyranyl, furanyl, pyrrolidyl, piperidinyl, thiolanyl, oxazolanyl, thiazolanyl, diazolanyl, piperazinyl, morpholino or thiomorpholino ring;
any lower alkyl group substituent on any of the substituents in R3-R6 which contain such a moiety can be optionally substituted with one or more of hydroxy, amino, lower monoalkylamino, lower dialkylamino, N-pyrrolidyl, N-piperidinyl, N-pyridinium, N-morpholino, N-thiomorpholino or N-piperazino groups;
R8 is hydrogen, lower alkyl (1-4 carbon atoms), amino or mono or dialkyl (1-4 carbon atoms) amino;
with the proviso that R3 cannot be either OH or SH;
with the further proviso that at least one of the R3 and R4 or R4 and R6 substituents must be other than hydrogen, halogen, lower alkyl (1-4 carbon atoms) or lower alkoxy (1-4 carbon atoms);
optionally if any of the substituents R1, R2, R3, R4 or R6 have chiral centers, then all stereoisomers thereof both as racemic and/or diastereoisomeric mixtures are included;
or a pharmaceutical salt or hydrate thereof.
28. A process for the preparation of 4-(3-bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine and pharmaceutically acceptable salts thereof which comprises:
Step (a) reaction of 5-amino-2-fluoropyridine with t-Boc anhydride to afford 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine;
Step (b) reaction of 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine sequentially with n-butyllithium and carbon dioxide to afford 5-[N-tert-butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid;
Step (c) reaction of 5-[N-tert-butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid with trifluoroacetic acid to afford 5-amino-2-fluoropyridine-4-carboxylic acid;
Step (d) reaction of 5-amino-2-fluoropyridine-4-carboxylic acid with formamide to afford 6-fluoro-3H-pyrido[3,4-d]pyrimidine-4-one;
Step (e) reaction of 6-fluoro-3H-pyrido[3,4-d]pyrimidine-4-one with phosphorus oxychloride to afford 4-chloro-6-fluoropyrido[3,4-d]pyrimidine;
Step (f) reaction of 4-chloro-6-fluoropyrido[3,4-d]pyrimidine with 3-bromoaniline to afford 4-(3-bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine;
Step (g) reaction of 4-(3bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine with methylamine to afford 4-(3-bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine; and
Step (h) if desired converting 4-(3-bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine to a corresponding pharmaceutically acceptable salt by conventional means, and if so desired converting the corresponding pharmaceutically acceptable salt to 4-(3-bromoanilino)-6-methylaminopyrido[3,4-d]pyrimidine by conventional means.
29. A compound which is 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine.
30. A compound which is 5-[N-(tert-butoxycarbonyl)amino]-2-fluoropyridine-4-carboxylic acid.
31. A compound which is 5-amino-2-fluoropyridine-4-carboxylic acid.
32. A compound which is 6-fluoro-3H-pyrido [3,4-d]pyrimidine-4-one.
33. A compound which is 4-chloro-6 fluoropyrido[3,4-d]pyrimidine.
34. A compound which is 4-(3-bromoanilino)-6-fluoropyrido[3,4-d]pyrimidine.
US09/824,606 1994-01-25 2001-04-02 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family Expired - Fee Related US6455534B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/824,606 US6455534B2 (en) 1994-01-25 2001-04-02 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US18674594A 1994-01-25 1994-01-25
US18673594A 1994-01-25 1994-01-25
US08/358,351 US5654307A (en) 1994-01-25 1994-12-23 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US08/811,797 US6084095A (en) 1994-01-25 1997-03-06 Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/191,163 US6265410B1 (en) 1994-01-25 1998-11-13 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/824,606 US6455534B2 (en) 1994-01-25 2001-04-02 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/191,163 Division US6265410B1 (en) 1994-01-25 1998-11-13 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Publications (2)

Publication Number Publication Date
US20010027197A1 true US20010027197A1 (en) 2001-10-04
US6455534B2 US6455534B2 (en) 2002-09-24

Family

ID=27392143

Family Applications (5)

Application Number Title Priority Date Filing Date
US08/811,797 Expired - Fee Related US6084095A (en) 1994-01-25 1997-03-06 Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/183,190 Expired - Fee Related US6521620B1 (en) 1994-01-25 1998-10-30 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/191,163 Expired - Fee Related US6265410B1 (en) 1994-01-25 1998-11-13 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/824,606 Expired - Fee Related US6455534B2 (en) 1994-01-25 2001-04-02 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US10/201,808 Expired - Fee Related US6713484B2 (en) 1994-01-25 2002-07-24 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/811,797 Expired - Fee Related US6084095A (en) 1994-01-25 1997-03-06 Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/183,190 Expired - Fee Related US6521620B1 (en) 1994-01-25 1998-10-30 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US09/191,163 Expired - Fee Related US6265410B1 (en) 1994-01-25 1998-11-13 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/201,808 Expired - Fee Related US6713484B2 (en) 1994-01-25 2002-07-24 Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Country Status (21)

Country Link
US (5) US6084095A (en)
EP (1) EP0742717A1 (en)
JP (1) JPH09508127A (en)
CN (2) CN1493291A (en)
AU (1) AU686334B2 (en)
BG (1) BG63245B1 (en)
CA (1) CA2177372A1 (en)
CZ (1) CZ197096A3 (en)
FI (3) FI114213B (en)
GE (1) GEP20012376B (en)
HR (1) HRP950034A2 (en)
HU (1) HU221741B1 (en)
IL (1) IL112249A (en)
MD (1) MD1632G2 (en)
NO (1) NO309892B1 (en)
NZ (1) NZ281011A (en)
PL (1) PL179132B1 (en)
RO (1) RO117257B1 (en)
SK (1) SK89496A3 (en)
TJ (1) TJ381B (en)
WO (1) WO1995019774A1 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007115822A1 (en) * 2006-04-07 2007-10-18 Develogen Aktiengesellschaft Thienopyrimidines having mnkl /mnk2 inhibiting activity for pharmaceutical compositions
US20090312319A1 (en) * 2008-01-04 2009-12-17 Intellikine Certain chemical entities, compositions and methods
US20100105708A1 (en) * 2006-07-10 2010-04-29 Develogen Aktiengesellschaft Pyrrolopyrimidines for pharmaceutical compositions
US20100143341A1 (en) * 2005-06-22 2010-06-10 Develogen Aktiengesellschaft Thienopyrimidines for pharmaceutical compositions
US20110046165A1 (en) * 2008-01-04 2011-02-24 Pingda Ren Certain chemical entitles, compositions and methods
US20110124641A1 (en) * 2008-03-14 2011-05-26 Pingda Ren Benzothiazole kinase inhibitors and methods of use
US20110130711A1 (en) * 2009-11-19 2011-06-02 Follica, Inc. Hair growth treatment
US20110130706A1 (en) * 2009-11-19 2011-06-02 Follica, Inc. Sequential body surface treatment
US20110212102A1 (en) * 2010-02-26 2011-09-01 Boehringer Ingelheim International Gmbh Heterocycloalkyl-containing thienopyrimidines for pharmaceutical compositions
US20110212103A1 (en) * 2010-02-26 2011-09-01 Boehringer Ingelheim International Gmbh Thienopyrimidines containing a substituted alkyl group for pharmaceutical compositions
US20110217311A1 (en) * 2010-02-26 2011-09-08 Boehringer Ingelheim International Gmbh Cycloalkyl containing thienopyrimidines for pharmaceutical compositions
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
US8486953B2 (en) 2008-08-26 2013-07-16 Boehringer Ingelheim International Gmbh Thienopyrimidines for pharmaceutical compositions
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US8901133B2 (en) 2010-11-10 2014-12-02 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US9295673B2 (en) 2011-02-23 2016-03-29 Intellikine Llc Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies

Families Citing this family (407)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7772432B2 (en) 1991-09-19 2010-08-10 Astrazeneca Ab Amidobenzamide derivatives which are useful as cytokine inhibitors
EP0682027B1 (en) * 1994-05-03 1997-10-15 Novartis AG Pyrrolopyrimidine derivatives with antiproliferative action
MX9702307A (en) * 1994-09-29 1998-04-30 Novartis Ag PYRROLO[2,3-d]PYRIMIDINES AND THEIR USE.
TW321649B (en) * 1994-11-12 1997-12-01 Zeneca Ltd
GB9424233D0 (en) * 1994-11-30 1995-01-18 Zeneca Ltd Quinazoline derivatives
DK0819129T3 (en) * 1995-04-03 2000-10-23 Novartis Ag Pyrazole derivatives and process for their preparation
GB9508535D0 (en) * 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivative
AU5343096A (en) * 1995-04-27 1996-11-18 Zeneca Limited Quinazoline derivatives
GB9508565D0 (en) * 1995-04-27 1995-06-14 Zeneca Ltd Quiazoline derivative
GB9508537D0 (en) * 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
GB9508538D0 (en) * 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
US6395733B1 (en) 1995-06-07 2002-05-28 Pfizer Inc Heterocyclic ring-fused pyrimidine derivatives
HUP9900330A3 (en) * 1995-07-06 2001-08-28 Novartis Ag Pyrrolopyrimidines and processes for the preparation thereof
AR004010A1 (en) 1995-10-11 1998-09-30 Glaxo Group Ltd HETERO CYCLIC COMPOUNDS
GB9624482D0 (en) 1995-12-18 1997-01-15 Zeneca Phaema S A Chemical compounds
ZA9756B (en) * 1996-01-16 1997-07-17 Warner Lambert Co Process for preparing 4,6-disubstituted pyrido[3,4-d]-pyrimidines
PT888349E (en) * 1996-01-23 2002-10-31 Novartis Ag PYRROLOPYRIMIDINES AND PROCESSES FOR THEIR PREPARATION
US5760041A (en) * 1996-02-05 1998-06-02 American Cyanamid Company 4-aminoquinazoline EGFR Inhibitors
CZ291386B6 (en) 1996-02-13 2003-02-12 Zeneca Limited Quinazoline derivatives functioning as VEGF inhibitors, process of their preparation and pharmaceutical preparations in which they are comprised
GB9603095D0 (en) 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline derivatives
GB9603097D0 (en) * 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline compounds
GB9604361D0 (en) * 1996-02-29 1996-05-01 Pharmacia Spa 4-Substituted pyrrolopyrimidine compounds as tyrosine kinase inhibitors
NZ331191A (en) 1996-03-05 2000-03-27 Zeneca Ltd 4-anilinoquinazoline derivatives and pharmaceutical compositions thereof
DE19608653A1 (en) * 1996-03-06 1997-09-11 Thomae Gmbh Dr K Pyrimido [5,4-d] pyrimidines, medicaments containing these compounds, their use and processes for their preparation
DE19629652A1 (en) * 1996-03-06 1998-01-29 Thomae Gmbh Dr K 4-Amino-pyrimidine derivatives, medicaments containing these compounds, their use and processes for their preparation
ES2174250T5 (en) 1996-04-12 2010-04-21 Warner-Lambert Company Llc IRREVERSIBLE THYROSINE KINASE INHIBITORS.
KR100545274B1 (en) * 1996-04-12 2006-04-12 워너-램버트 캄파니 엘엘씨 Irreversible Inhibitors of Tyrosine Kinases
GB9607729D0 (en) * 1996-04-13 1996-06-19 Zeneca Ltd Quinazoline derivatives
GB9613021D0 (en) * 1996-06-21 1996-08-28 Pharmacia Spa Bicyclic 4-aralkylaminopyrimidine derivatives as tyrosine kinase inhibitors
DE69718472T2 (en) * 1996-07-13 2003-11-06 Glaxo Group Ltd BICYCLIC HETEROAROMATIC COMPOUNDS AS PROTEIN TYROSINE KINASE INHIBITORS
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
WO1998002434A1 (en) 1996-07-13 1998-01-22 Glaxo Group Limited Fused heterocyclic compounds as protein tyrosine kinase inhibitors
BR9710808A (en) 1996-08-06 1999-08-17 Pfizer Bicyclic derivatives 6,6 or 6,7 containing substituted pyrite or pyrimido
CA2262421C (en) * 1996-08-23 2007-10-02 Novartis Ag Substituted pyrrolopyrimidines and processes for their preparation
ID18494A (en) * 1996-10-02 1998-04-16 Novartis Ag PIRAZOLA DISTRIBUTION IN THE SEQUENCE AND THE PROCESS OF MAKING IT
AU4779897A (en) * 1996-10-02 1998-04-24 Novartis Ag Fused pyrazole derivatives and processes for their preparation
JP4205168B2 (en) 1996-10-02 2009-01-07 ノバルティス アクチエンゲゼルシヤフト Pyrimidine derivatives and process for producing the same
BR9713552A (en) 1996-11-27 2000-01-25 Pfizer Condensed bicyclic pyrimidine derivatives
AU7890598A (en) * 1996-12-27 1998-07-31 Yoshitomi Pharmaceutical Industries, Ltd. Fused pyrimidine compounds and medicinal use thereof
US6002008A (en) * 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
TW458977B (en) * 1997-04-16 2001-10-11 Abbott Lab 6,7-disubstituted-4-aminopyrido[2,3-D] pyrimidine compounds
CN1252069A (en) * 1997-04-16 2000-05-03 艾博特公司 5,6,7-trisubstituted-4-aminopyrido [2,3-d] pyrimidine compounds
AU7108398A (en) * 1997-04-16 1998-11-11 Abbott Laboratories 5,7-disubstituted 4-aminopyrido{2,3-d}pyrimidine compounds and their use as adenosine kinase inhibitors
US5929080A (en) * 1997-05-06 1999-07-27 American Cyanamid Company Method of treating polycystic kidney disease
CA2288705C (en) * 1997-05-06 2008-03-18 American Cyanamid Company Use of quinazoline compounds for the treatment of polycystic kidney disease
ZA986729B (en) * 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitors of tyrosine kinases
ZA986732B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitiors of tyrosine kinases
US6251912B1 (en) 1997-08-01 2001-06-26 American Cyanamid Company Substituted quinazoline derivatives
US6323209B1 (en) 1997-11-06 2001-11-27 American Cyanamid Company Method of treating or inhibiting colonic polyps
CN101328186A (en) 1997-11-11 2008-12-24 辉瑞产品公司 Thienopyrimidine and thienopyridine derivatives useful as anticancer agents
RS49779B (en) * 1998-01-12 2008-06-05 Glaxo Group Limited, Byciclic heteroaromatic compounds as protein tyrosine kinase inhibitors
US6030969A (en) * 1998-04-09 2000-02-29 Abbott Laboratories 5,6,7-trisubstituted-4-aminopyrido[2,3-D] pyrimidine compounds
CN1311678A (en) * 1998-04-30 2001-09-05 巴斯福股份公司 Substituted tricyclic pyrazole derivatives with protein kinase activity
HUP0102295A3 (en) 1998-05-15 2002-11-28 Astrazeneca Ab Benzamide derivatives for the treatment of diseases mediated by cytokines
PL344248A1 (en) 1998-05-26 2001-10-22 Warner Lambert Co Bicyclic pyrimidines and bicyclic 3,4-dihydropyrimidines as inhibitors of cellular proliferation
EP1801112A1 (en) * 1998-05-26 2007-06-27 Warner-Lambert Company LLC Bicyclic pyrimidines and bicyclic 3,4-dihydropyrimidines as inhibitors of cellular proliferation
US20040044012A1 (en) * 1998-05-26 2004-03-04 Dobrusin Ellen Myra Bicyclic pyrimidines and bicyclic 3,4-dihydropyrimidines as inhibitors of cellular proliferation
US6878716B1 (en) 1998-06-02 2005-04-12 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 receptor and uses thereof
AP1411A (en) 1998-06-02 2005-05-10 Osi Pharm Inc Pyrrolo[2,3d] pyrimidine compositions and their use.
US6686366B1 (en) 1998-06-02 2004-02-03 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
AU3951899A (en) 1998-06-19 2000-01-05 Pfizer Products Inc. Pyrrolo(2,3-d)pyrimidine compounds
PA8474101A1 (en) 1998-06-19 2000-09-29 Pfizer Prod Inc PYROLEUM [2,3-D] PIRIMIDINE COMPOUNDS
EP1105394A1 (en) 1998-08-21 2001-06-13 Du Pont Pharmaceuticals Company ISOXAZOLO 4,5-d]PYRIMIDINES AS CRF ANTAGONISTS
WO2000018738A1 (en) 1998-09-25 2000-04-06 Astrazeneca Ab Benzamide derivatives and their use as cytokine inhibitors
NZ510580A (en) 1998-09-29 2003-03-28 American Cyanamid Co Substituted 3-cyanoquinolines useful as protein tyrosine kinases inhibitors
US6288082B1 (en) 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
US6297258B1 (en) 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
PT1117653E (en) 1998-10-01 2003-06-30 Astrazeneca Ab QUINOLINE AND QUINAZOLINE DERIVATIVES AND THEIR USE AS CYTOSINE MEDIUM-INHIBITORS
DK1119567T3 (en) 1998-10-08 2005-07-25 Astrazeneca Ab quinazoline
WO2000023444A1 (en) * 1998-10-21 2000-04-27 Abbott Laboratories 5,7-disubstituted-4-aminopyrido[2,3-d]pyrimidine compounds
JP3270834B2 (en) 1999-01-27 2002-04-02 ファイザー・プロダクツ・インク Heteroaromatic bicyclic derivatives useful as anticancer agents
UA71945C2 (en) 1999-01-27 2005-01-17 Pfizer Prod Inc Substituted bicyclic derivatives being used as anticancer agents
US7008945B1 (en) 1999-03-17 2006-03-07 Astrazeneca Ab Amide derivatives
GB9906566D0 (en) 1999-03-23 1999-05-19 Zeneca Ltd Chemical compounds
ID30460A (en) 1999-04-15 2001-12-06 Bristol Myers Squibb Co INHIBITORS, INHIBITORS, THYROCIN CINASE PROTEIN
US7125875B2 (en) 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
ES2295035T3 (en) 1999-07-09 2008-04-16 Glaxo Group Limited ANILINOQUINAZOLINAS AS INHIBITORS OF PROTEIN TIROSINA QUINASA.
US6933299B1 (en) 1999-07-09 2005-08-23 Smithkline Beecham Corporation Anilinoquinazolines as protein tyrosine kinase inhibitors
US6432979B1 (en) 1999-08-12 2002-08-13 American Cyanamid Company Method of treating or inhibiting colonic polyps and colorectal cancer
CN101219145A (en) 1999-11-05 2008-07-16 阿斯特拉曾尼卡有限公司 Quinazoline derivatives as VEGF inhibitors
US7160890B2 (en) 1999-12-02 2007-01-09 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
PT1235830E (en) 1999-12-10 2004-04-30 Pfizer Prod Inc PYRIMIDINE-2,3-D | PYRIMIDINE COMPOUNDS AS PROTEIN KINASE INHIBITORS
US7235551B2 (en) 2000-03-02 2007-06-26 Smithkline Beecham Corporation 1,5-disubstituted-3,4-dihydro-1h-pyrimido[4,5-d]pyrimidin-2-one compounds and their use in treating csbp/p38 kinase mediated diseases
ATE277933T1 (en) 2000-06-06 2004-10-15 Pfizer Prod Inc THIOPHENE COMPOUNDS FOR USE AS ANTICANCER AGENTS
PL359563A1 (en) 2000-06-26 2004-08-23 Pfizer Products Inc. Pyrrolo[2,3-d]pyrimidine compounds as immunosuppressive agents
EP1170011A1 (en) * 2000-07-06 2002-01-09 Boehringer Ingelheim International GmbH Novel use of inhibitors of the epidermal growth factor receptor
ATE399766T1 (en) 2000-10-20 2008-07-15 Eisai R&D Man Co Ltd AROMATIC HETEROCYCLES CONTAINING NITROGEN
US6503914B1 (en) 2000-10-23 2003-01-07 Board Of Regents, The University Of Texas System Thienopyrimidine-based inhibitors of the Src family
IL155367A0 (en) 2000-10-23 2003-12-23 Smithkline Beecham Corp NOVEL 2,4,8-TRISUBSTITUTED-8h-PYRIDO[2,3,-d]PYRIMIDIN-7-ONE COMPOUNDS, PHARMACEUTICAL COMPOSITIONS COMPRISING THE SAME, PROCESSES FOR THE PREPARATION THEREOF, AND USE THEREOF IN THE PREPARATION OF MEDICAMENTS FOR TREATING CSBP/p38 KINASE MEDIATED DISEASES
US6673802B2 (en) 2000-12-01 2004-01-06 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6680324B2 (en) 2000-12-01 2004-01-20 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 receptors and uses thereof
DE60212487T2 (en) 2001-04-13 2006-12-21 Pfizer Products Inc., Groton Bicyclic substituted 4-aminopyridopyrimidine derivatives
JP4307843B2 (en) * 2001-04-19 2009-08-05 アストラゼネカ アクチボラグ Quinazoline derivatives
US7301023B2 (en) 2001-05-31 2007-11-27 Pfizer Inc. Chiral salt resolution
US6995171B2 (en) 2001-06-21 2006-02-07 Agouron Pharmaceuticals, Inc. Bicyclic pyrimidine and pyrimidine derivatives useful as anticancer agents
DE10154540A1 (en) * 2001-11-07 2003-05-22 Cellcontrol Biomedical Lab Ag Procedure for predicting or predicting the effectiveness of tumor treatment
US20030166509A1 (en) * 2001-11-20 2003-09-04 Advanced Inhalation Research, Inc. Compositions for sustained action product delivery and methods of use thereof
AU2002360436A1 (en) 2001-11-30 2003-06-17 Osi Pharmaceuticals, Inc. 2-aryl pyrrologpyrimidines for a1 and a3 receptors
BR0214499A (en) * 2001-12-12 2005-05-10 Pfizer Prod Inc Quinazoline derivatives for the treatment of abnormal cell growth
US20030229067A1 (en) 2001-12-20 2003-12-11 Arlindo Castelhano Pyrrolopyrimidine A2b selective antagonist compounds, their synthesis and use
CA2471059C (en) 2001-12-20 2011-04-26 Osi Pharmaceuticals, Inc. Pyrimidine a2b selective antagonist compounds, their synthesis and use
HUP0500200A2 (en) 2002-01-17 2005-07-28 Neurogen Corporation Substituted quinazolin-4-ylamine analogues as modulators of capsaicin and pharmaceutical compositions thereof
US7078409B2 (en) * 2002-03-28 2006-07-18 Beta Pharma, Inc. Fused quinazoline derivatives useful as tyrosine kinase inhibitors
US7176310B1 (en) 2002-04-09 2007-02-13 Ucb Sa Pyrimidinecarboxamide derivatives and their use as anti-inflammatory agents
EP1499320B1 (en) 2002-04-19 2007-08-22 Smithkline Beecham Corporation Novel compounds
JP2005536486A (en) * 2002-07-09 2005-12-02 アストラゼネカ アクチボラグ Quinazoline derivatives for use in the treatment of cancer
US7196090B2 (en) * 2002-07-25 2007-03-27 Warner-Lambert Company Kinase inhibitors
EP1553947A4 (en) * 2002-10-21 2006-11-29 Bristol Myers Squibb Co Quinazolinones and derivatives thereof as factor xa inhibitors
MXPA05005576A (en) 2002-11-26 2005-07-27 Pfizer Prod Inc Method of treatment of transplant rejection.
JP5129957B2 (en) * 2003-07-03 2013-01-30 ミリアド ジェネティクス, インコーポレイテッド 4-Arylamino-quinazolines as caspase activators and inducers of apoptosis
US8309562B2 (en) 2003-07-03 2012-11-13 Myrexis, Inc. Compounds and therapeutical use thereof
AU2004275842B2 (en) 2003-09-26 2010-09-02 Exelixis Inc. c-MET modulators and methods of use
US7419978B2 (en) 2003-10-22 2008-09-02 Bristol-Myers Squibb Company Phenyl-aniline substituted bicyclic compounds useful as kinase inhibitors
GB0324790D0 (en) 2003-10-24 2003-11-26 Astrazeneca Ab Amide derivatives
CN101337930B (en) 2003-11-11 2010-09-08 卫材R&D管理有限公司 Urea derivative preparation process
WO2005058913A1 (en) 2003-12-18 2005-06-30 Janssen Pharmaceutica N.V. Pyrido- and pyrimidopyrimidine derivatives as anti- proliferative agents
WO2005095419A1 (en) * 2004-04-01 2005-10-13 Takeda Pharmaceutial Company Limited Thiazolopyrimidine derivative
MEP8409A (en) * 2004-06-02 2011-12-20 Fused heterocyclic compound
JP5368701B2 (en) 2004-07-02 2013-12-18 エクセリクシス、インコーポレイテッド c-Met Modulator and Method of Use
JP2008506714A (en) 2004-07-16 2008-03-06 サネシス ファーマシューティカルズ, インコーポレイテッド Thienopyrimidines useful as aurora kinase inhibitors
US7300940B2 (en) 2004-08-04 2007-11-27 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Integrin α-v β-3 antagonists for use in imaging and therapy
AU2005283422C1 (en) 2004-09-17 2017-02-02 Eisai R & D Management Co., Ltd. Medicinal composition
JO3088B1 (en) * 2004-12-08 2017-03-15 Janssen Pharmaceutica Nv Macrocyclic Quinazoline derivatives and their use as MTKI
NI200700147A (en) 2004-12-08 2019-05-10 Janssen Pharmaceutica Nv QUINAZOLINE DERIVATIVES KINE INHIBITORS TARGETING MULTIP
ATE501148T1 (en) 2004-12-14 2011-03-15 Astrazeneca Ab PYRAZOLOPYRIMIDINE COMPOUNDS AS ANTI-TUMOR AGENTS
US20090264415A2 (en) * 2004-12-30 2009-10-22 Steven De Jonghe Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for medical treatment
GB0428475D0 (en) * 2004-12-30 2005-02-02 4 Aza Bioscience Nv Pyrido(3,2-D)pyrimidine derivatives and pharmaceutical compositions useful as medicines for the treatment of autoimmune disorders
EP1833482A4 (en) 2005-01-03 2011-02-16 Myriad Genetics Inc Compounds and therapeutical use thereof
US8258145B2 (en) 2005-01-03 2012-09-04 Myrexis, Inc. Method of treating brain cancer
US20090155247A1 (en) * 2005-02-18 2009-06-18 Ashkenazi Avi J Methods of Using Death Receptor Agonists and EGFR Inhibitors
US20060188498A1 (en) * 2005-02-18 2006-08-24 Genentech, Inc. Methods of using death receptor agonists and EGFR inhibitors
US20080096905A1 (en) 2005-03-25 2008-04-24 Glaxo Group Limited Process For Preparing Pyrido[2,3-D]Pyrimidin-7-One And 3,4-Dihydropyrimido{4,5-D}Pyrimidin-2(1H)-One Derivatives
WO2006110298A2 (en) 2005-03-25 2006-10-19 Glaxo Group Limited 8-alkyl/aryl-4-aryl-2-n- (alkylamino)-n'-substituted-n'-cyanoguanidino-8h-pyrido[2,3-d]pyrimidin-7-one compounds and use thereof
ATE517897T1 (en) 2005-03-25 2011-08-15 Tibotec Pharm Ltd HETEROBICYCLIC INHIBITORS OF HVC
PE20100741A1 (en) 2005-03-25 2010-11-25 Glaxo Group Ltd COMPOUNDS DERIVED FROM 3,4-DIHYDROPYRIMIDE [4,5-d] PYRIMIDIN-2 (1H) -ONE AS KINASE INHIBITORS p38
PE20061351A1 (en) 2005-03-25 2007-01-14 Glaxo Group Ltd 8H-PYRID [2,3-d] PYRIMIDIN-7-ONA 2,4,8-TRISUSTITUTED COMPOUNDS AS CSBP / RK / p38 KINASE INHIBITORS
AR056347A1 (en) 2005-05-12 2007-10-03 Tibotec Pharm Ltd USE OF PTERIDINE COMPOUNDS TO MANUFACTURE PHARMACEUTICAL MEDICINES AND COMPOSITIONS
AR054122A1 (en) 2005-05-12 2007-06-06 Tibotec Pharm Ltd PIRIDO [2,3-D] USEFUL PYRIMIDES AS HCV INHIBITORS, AND METHODS FOR THE PREPARATION OF THE SAME
CA2612788A1 (en) * 2005-06-24 2006-12-28 Steven Cesar Alfons De Jonghe Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for treating hepatitis c
TW200740820A (en) * 2005-07-05 2007-11-01 Takeda Pharmaceuticals Co Fused heterocyclic derivatives and use thereof
AU2006272876A1 (en) 2005-07-22 2007-02-01 Sunesis Pharmaceuticals, Inc. Pyrazolo pyrimidines useful as aurora kinase inhibitors
EP1925676A4 (en) 2005-08-02 2010-11-10 Eisai R&D Man Co Ltd Method for assay on the effect of vascularization inhibitor
JP2009507849A (en) * 2005-09-09 2009-02-26 ブリストル−マイヤーズ スクイブ カンパニー Acyclic IKur inhibitor
TW200730527A (en) * 2005-12-02 2007-08-16 Takeda Pharmaceuticals Co Fused heterocyclic compound
JP2009521480A (en) 2005-12-21 2009-06-04 アボット・ラボラトリーズ Antiviral compounds
ES2378473T3 (en) 2005-12-21 2012-04-12 Abbott Laboratories Antiviral compounds
US7915411B2 (en) 2005-12-21 2011-03-29 Abbott Laboratories Anti-viral compounds
EP2345652A1 (en) 2005-12-21 2011-07-20 Abbott Laboratories Antiviral compounds
CN101443009A (en) 2006-05-18 2009-05-27 卫材R&D管理有限公司 Antitumor agent for thyroid cancer
DE102006029573A1 (en) * 2006-06-22 2007-12-27 Bayer Schering Pharma Ag New quinazolines as kinase inhibitors
CN101100466B (en) 2006-07-05 2013-12-25 天津和美生物技术有限公司 Non-reversible protein tyrosine phosphorus acylase inhibitor and its preparation and application
AU2007274284B2 (en) 2006-07-13 2012-04-26 Janssen Pharmaceutica N.V. Mtki quinazoline derivatives
US8338435B2 (en) * 2006-07-20 2012-12-25 Gilead Sciences, Inc. Substituted pyrido(3,2-D) pyrimidines and pharmaceutical compositions for treating viral infections
US8865737B2 (en) 2006-08-28 2014-10-21 Eisai R&D Management Co., Ltd. Antitumor agent for undifferentiated gastric cancer
ATE522625T1 (en) 2006-09-12 2011-09-15 Genentech Inc METHODS AND COMPOSITIONS FOR THE DIAGNOSIS AND TREATMENT OF LUNG CANCER USING PDGFRA, KIT OR KDG GENE AS GENETIC MARKERS
KR20090090336A (en) * 2006-12-12 2009-08-25 다케다 야쿠힌 고교 가부시키가이샤 Fused heterocyclic compound
WO2008133753A2 (en) 2006-12-20 2008-11-06 Abbott Laboratories Anti-viral compounds
WO2008077649A1 (en) * 2006-12-26 2008-07-03 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines useful for treating viral infectons
TW200840584A (en) * 2006-12-26 2008-10-16 Gilead Sciences Inc Pyrido(3,2-d)pyrimidines useful for treating viral infections
WO2008077650A1 (en) * 2006-12-26 2008-07-03 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines useful for treating viral infections
JP5319306B2 (en) 2007-01-29 2013-10-16 エーザイ・アール・アンド・ディー・マネジメント株式会社 Composition for treatment of undifferentiated gastric cancer
CA2679694A1 (en) * 2007-03-13 2008-09-18 Merck & Co., Inc. Inhibitors of janus kinases and/or 3-phosphoinositide-dependent protein kinase-1
DK2185562T3 (en) 2007-07-27 2016-02-22 Janssen Pharmaceutica Nv PYRROLOPYRIMIDINES SUITABLE FOR TREATING PROLIFERATIVE DISEASES
KR101513326B1 (en) 2007-11-09 2015-04-17 에자이 알앤드디 매니지먼트 가부시키가이샤 Combination of anti-angiogenic substance and anti-tumor platinum complex
WO2009067543A2 (en) * 2007-11-19 2009-05-28 The Regents Of The University Of Colorado Treatment of histone deacetylase mediated disorders
CA2707046A1 (en) * 2007-11-28 2009-06-11 Nathanael S. Gray Small molecule myristate inhibitors of bcr-abl and methods of use
CN101492445A (en) * 2008-01-22 2009-07-29 孙飘扬 Heteroaromatic compound, preparation method and uses thereof
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
PE20091580A1 (en) * 2008-03-12 2009-11-05 Takeda Pharmaceutical FUSED HETEROCYCLIC COMPOUND
CN101575333B (en) * 2008-05-09 2011-06-22 和记黄埔医药(上海)有限公司 Quinazoline derivatives and medical application thereof
US8536187B2 (en) * 2008-07-03 2013-09-17 Gilead Sciences, Inc. 2,4,6-trisubstituted pyrido(3,2-d)pyrimidines useful for treating viral infections
BRPI0917459B1 (en) 2008-08-20 2017-09-12 Zoetis Services Llc N-METHYL-1- [TRANS-4- [METHYL (7H-PYRROL [2,3-D] PYRIDOL [2,3-D] PYRIMIDINE COMPOUNDS, USE OF THESE IN THERAPY AND CRYSTALLINE AS N-METHYL-PYRIMIDIN -4-YL) AMINO] CYCLOHEXYL} METHANOSULPHONAMIDE
AR073501A1 (en) * 2008-09-08 2010-11-10 Boehringer Ingelheim Int PYRIMID DERIVATIVES [5,4-D] PYRIMIDINE INHIBITORS OF THYROSINOQUINASE
CA2742986C (en) 2008-11-07 2015-03-31 Triact Therapeutics, Inc. Use of catecholic butane derivatives in cancer therapy
CN102264745B (en) * 2008-11-10 2015-07-22 财团法人卫生研究院 Fused bicyclic and tricyclic pyrimidine compounds as tyrosine kinase inhibitors
MX2011005720A (en) * 2008-12-01 2011-06-17 Merck Patent Gmbh 2, 5-diamino-substituted pyrido [4, 3-d] pyrimidines as autotaxin inhibitors against cancer.
KR20210151988A (en) 2009-01-16 2021-12-14 엑셀리시스, 인코포레이티드 Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
CA2752265A1 (en) * 2009-02-17 2010-08-26 Boehringer Ingelheim International Gmbh Pyrimido [5,4-d] pyrimidine derivatives for the inhibition of tyrosine kinases
US20120189641A1 (en) 2009-02-25 2012-07-26 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
EP2400990A2 (en) 2009-02-26 2012-01-04 OSI Pharmaceuticals, LLC In situ methods for monitoring the emt status of tumor cells in vivo
WO2010099138A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
JP2012519282A (en) 2009-02-27 2012-08-23 オーエスアイ・ファーマシューティカルズ,エルエルシー Methods for identifying mesenchymal tumor cells or agents that inhibit their production
US8642834B2 (en) 2009-02-27 2014-02-04 OSI Pharmaceuticals, LLC Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
JP2012520893A (en) 2009-03-18 2012-09-10 オーエスアイ・ファーマシューティカルズ,エルエルシー Combination cancer treatment comprising administration of an EGFR inhibitor and an IGF-1R inhibitor
UA106070C2 (en) 2009-03-20 2014-07-25 Дженентек, Інк. Antibody that specifically binds to egfr and her3
UA108618C2 (en) 2009-08-07 2015-05-25 APPLICATION OF C-MET-MODULATORS IN COMBINATION WITH THEMOSOLOMID AND / OR RADIATION THERAPY FOR CANCER TREATMENT
HUE032983T2 (en) 2009-09-03 2017-11-28 Bristol Myers Squibb Co Quinazolines as potassium ion channel inhibitors
JP2013510871A (en) 2009-11-12 2013-03-28 ジェネンテック, インコーポレイテッド How to increase the density of dendritic spines
WO2011058164A1 (en) 2009-11-13 2011-05-19 Pangaea Biotech, S.A. Molecular biomarkers for predicting response to tyrosine kinase inhibitors in lung cancer
RU2012132278A (en) 2010-01-12 2014-02-20 Ф. Хоффманн-Ля Рош Аг TRICYCLIC HETEROCYCLIC COMPOUNDS CONTAINING THEIR COMPOSITIONS AND WAYS OF THEIR APPLICATION
SG183333A1 (en) 2010-02-18 2012-09-27 Genentech Inc Neuregulin antagonists and use thereof in treating cancer
CN102869359A (en) 2010-03-17 2013-01-09 弗·哈夫曼-拉罗切有限公司 Imidazopyridine compounds, compositions and methods of use
EP2558864A1 (en) 2010-04-16 2013-02-20 Genentech, Inc. Fox03a as predictive biomarker for pi3k/akt kinase pathway inhibitor efficacy
WO2011162343A1 (en) 2010-06-25 2011-12-29 エーザイ・アール・アンド・ディー・マネジメント株式会社 Antitumor agent using compounds having kinase inhibitory effect in combination
DE102010033690A1 (en) * 2010-08-06 2012-02-09 Saltigo Gmbh Process for the preparation of aminoarylalkyl compounds
DK2612151T3 (en) 2010-08-31 2017-10-02 Genentech Inc BIOMARKETS AND METHODS OF TREATMENT
BR112013006016A2 (en) 2010-09-15 2016-06-07 Hoffmann La Roche azabenzothiazole compounds, compositions and methods of use
KR20140009259A (en) 2010-11-19 2014-01-22 에프. 호프만-라 로슈 아게 Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
CN102532107B (en) * 2010-12-20 2014-03-12 天津药物研究院 4-substituted aniline-7-substituted alcoxylhomopiperazine-quinazoline derivatives, and preparation method and application thereof
EP2468883A1 (en) 2010-12-22 2012-06-27 Pangaea Biotech S.L. Molecular biomarkers for predicting response to tyrosine kinase inhibitors in lung cancer
WO2012085176A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Tricyclic pyrazinone compounds, compositions and methods of use thereof as janus kinase inhibitors
US9134297B2 (en) 2011-01-11 2015-09-15 Icahn School Of Medicine At Mount Sinai Method and compositions for treating cancer and related methods
US9139590B2 (en) 2011-02-04 2015-09-22 Duquesne University Of The Holy Spirit Bicyclic and tricyclic pyrimidine tyrosine kinase inhibitors with antitubulin activity and methods of treating a patient
EP2492688A1 (en) 2011-02-23 2012-08-29 Pangaea Biotech, S.A. Molecular biomarkers for predicting response to antitumor treatment in lung cancer
WO2012129145A1 (en) 2011-03-18 2012-09-27 OSI Pharmaceuticals, LLC Nscle combination therapy
US8962650B2 (en) 2011-04-18 2015-02-24 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
EP2702173A1 (en) 2011-04-25 2014-03-05 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
JP6038128B2 (en) 2011-06-03 2016-12-07 エーザイ・アール・アンド・ディー・マネジメント株式会社 A biomarker for predicting and evaluating the reactivity of thyroid and renal cancer subjects to lenvatinib compounds
WO2013007768A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Tricyclic heterocyclic compounds, compositions and methods of use thereof as jak inhibitors
WO2013007765A1 (en) 2011-07-13 2013-01-17 F. Hoffmann-La Roche Ag Fused tricyclic compounds for use as inhibitors of janus kinases
CA2843499A1 (en) 2011-08-12 2013-02-21 F. Hoffmann-La Roche Ag Indazole compounds, compositions and methods of use
EP2744824A1 (en) 2011-08-17 2014-06-25 F.Hoffmann-La Roche Ag Neuregulin antibodies and uses thereof
WO2013026806A1 (en) * 2011-08-19 2013-02-28 Exonhit Sa Dyrk1 inhibitors and uses thereof
WO2013028818A1 (en) 2011-08-23 2013-02-28 Endo Pharmaceuticals Inc. Pyrimido- pyridazinone compounds and use thereof
CN104024432B (en) 2011-08-31 2017-02-22 基因泰克公司 Diagnostic Markers
EP2758397A1 (en) 2011-09-20 2014-07-30 F.Hoffmann-La Roche Ag Imidazopyridine compounds, compositions and methods of use
JP2014531213A (en) 2011-09-30 2014-11-27 ジェネンテック, インコーポレイテッド Diagnostic methylation markers for epithelial or mesenchymal phenotype and response to EGFR kinase inhibitors in tumors or tumor cells
AU2012346540C1 (en) 2011-11-30 2019-07-04 Genentech, Inc. ErbB3 mutations in cancer
ES2748590T3 (en) 2011-12-16 2020-03-17 Us Health Compounds and methods for the prevention and treatment of tumor metastases and tumorigenesis
SG11201406079TA (en) 2012-03-27 2014-10-30 Genentech Inc Diagnosis and treatments relating to her3 inhibitors
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
WO2013190089A1 (en) 2012-06-21 2013-12-27 Pangaea Biotech, S.L. Molecular biomarkers for predicting outcome in lung cancer
CA2887539C (en) 2012-11-29 2021-10-26 Merck Patent Gmbh Azaquinazoline carboxamide derivatives
MX2015004979A (en) 2012-12-21 2015-07-17 Eisai R&D Man Co Ltd Amorphous form of quinoline derivative, and method for producing same.
CA2900097A1 (en) 2013-02-22 2014-08-28 F. Hoffmann-La Roche Ag Methods of treating cancer and preventing drug resistance
EP2961412A4 (en) 2013-02-26 2016-11-09 Triact Therapeutics Inc Cancer therapy
US9925240B2 (en) 2013-03-06 2018-03-27 Genentech, Inc. Methods of treating and preventing cancer drug resistance
MX2015011898A (en) 2013-03-13 2016-05-05 Genentech Inc Pyrazolo compounds and uses thereof.
JP2016516046A (en) 2013-03-14 2016-06-02 ジェネンテック, インコーポレイテッド Methods for treating cancer and methods for preventing cancer drug resistance
KR20150127203A (en) 2013-03-14 2015-11-16 제넨테크, 인크. Combinations of a mek inhibitor compound with an her3/egfr inhibitor compound and methods of use
MX2015011899A (en) 2013-03-15 2016-05-05 Genentech Inc Methods of treating cancer and preventing cancer drug resistance.
EP2976085A1 (en) 2013-03-21 2016-01-27 INSERM - Institut National de la Santé et de la Recherche Médicale Method and pharmaceutical composition for use in the treatment of chronic liver diseases associated with a low hepcidin expression
JP6411379B2 (en) 2013-05-14 2018-10-24 エーザイ・アール・アンド・ディー・マネジメント株式会社 Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
MX2016002794A (en) 2013-09-05 2016-09-13 Genentech Inc Antiproliferative compounds.
CA2923667A1 (en) 2013-09-09 2015-03-12 Triact Therapeutics, Inc. Cancer therapy
TW201605857A (en) 2013-10-03 2016-02-16 赫孚孟拉羅股份公司 Therapeutic inhibitors of CDK8 and uses thereof
RU2016114074A (en) 2013-10-18 2017-11-23 Дженентек, Инк. ANTI-RSPO ANTIBODIES AND METHODS OF APPLICATION
KR20240017102A (en) 2013-12-17 2024-02-06 제넨테크, 인크. Methods of treating cancers using pd-1 axis binding antagonists and taxanes
CA2934028A1 (en) 2013-12-17 2015-06-25 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
EP2905024A1 (en) 2014-02-07 2015-08-12 Institut Quimic De Sarriá Cets, Fundació Privada Pyrido[2,3-d]pyrimidine-7(8H)-one derivatives for the treatment of infections caused by Flaviviridae
RU2016141385A (en) 2014-03-24 2018-04-28 Дженентек, Инк. CANCER TREATMENT WITH C-MET ANTAGONISTS AND THEIR CORRELATION WITH HGF EXPRESSION
PE20161571A1 (en) 2014-03-31 2017-02-07 Genentech Inc ANTI-OX40 ANTIBODIES AND METHODS OF USE
JP6588461B2 (en) 2014-03-31 2019-10-09 ジェネンテック, インコーポレイテッド Combination therapy comprising an anti-angiogenic agent and an OX40 binding agonist
WO2015156674A2 (en) 2014-04-10 2015-10-15 Stichting Het Nederlands Kanker Instituut Method for treating cancer
EP3825305A1 (en) 2014-08-28 2021-05-26 Eisai R&D Management Co., Ltd. Process for preparing lenvatinib
WO2016036873A1 (en) 2014-09-05 2016-03-10 Genentech, Inc. Therapeutic compounds and uses thereof
JP2017529358A (en) 2014-09-19 2017-10-05 ジェネンテック, インコーポレイテッド Use of CBP / EP300 inhibitors and BET inhibitors for the treatment of cancer
CN104370825B (en) * 2014-09-29 2017-04-19 人福医药集团股份公司 Substituted heterocyclic compound as kinase inhibitor and its preparation method and use
WO2016057924A1 (en) 2014-10-10 2016-04-14 Genentech, Inc. Pyrrolidine amide compounds as histone demethylase inhibitors
SG11201703448QA (en) 2014-11-03 2017-05-30 Genentech Inc Assays for detecting t cell immune subsets and methods of use thereof
MX2017005751A (en) 2014-11-03 2018-04-10 Genentech Inc Method and biomarkers for predicting efficacy and evaluation of an ox40 agonist treatment.
CA2963974A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Combination therapy comprising ox40 binding agonists and tigit inhibitors
MA40943A (en) 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS
EP3218376B1 (en) 2014-11-10 2019-12-25 Genentech, Inc. Bromodomain inhibitors and uses thereof
MA40940A (en) 2014-11-10 2017-09-19 Constellation Pharmaceuticals Inc SUBSTITUTED PYRROLOPYRIDINES USED AS BROMODOMA INHIBITORS
WO2016081384A1 (en) 2014-11-17 2016-05-26 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
US9763922B2 (en) 2014-11-27 2017-09-19 Genentech, Inc. Therapeutic compounds and uses thereof
WO2016106340A2 (en) 2014-12-23 2016-06-30 Genentech, Inc. Compositions and methods for treating and diagnosing chemotherapy-resistant cancers
CA2969830A1 (en) 2014-12-24 2016-06-30 Genentech, Inc. Therapeutic, diagnostic and prognostic methods for cancer of the bladder
EP3240908A2 (en) 2014-12-30 2017-11-08 F. Hoffmann-La Roche AG Methods and compositions for prognosis and treatment of cancers
CN107406414B (en) 2015-01-09 2022-04-19 基因泰克公司 (piperidin-3-yl) (naphthalen-2-yl) methanone derivatives as inhibitors of histone demethylase KDM2B for the treatment of cancer
WO2016112298A1 (en) 2015-01-09 2016-07-14 Genentech, Inc. Pyridazinone derivatives and their use in the treatment of cancer
EP3242875B1 (en) 2015-01-09 2022-02-23 Genentech, Inc. 4,5-dihydroimidazole derivatives and their use as histone demethylase (kdm2b) inhibitors
WO2016123391A1 (en) 2015-01-29 2016-08-04 Genentech, Inc. Therapeutic compounds and uses thereof
WO2016123387A1 (en) 2015-01-30 2016-08-04 Genentech, Inc. Therapeutic compounds and uses thereof
PT3263106T (en) 2015-02-25 2024-01-12 Eisai R&D Man Co Ltd Method for suppressing bitterness of quinoline derivative
MA41598A (en) 2015-02-25 2018-01-02 Constellation Pharmaceuticals Inc PYRIDAZINE THERAPEUTIC COMPOUNDS AND THEIR USES
AU2015384801B2 (en) 2015-03-04 2022-01-06 Eisai R&D Management Co., Ltd. Combination of a PD-1 antagonist and a VEGFR/FGFR/RET tyrosine kinase inhibitor for treating cancer
JP6955445B2 (en) 2015-04-07 2021-10-27 ジェネンテック, インコーポレイテッド Antigen binding complex with agonistic activity and how to use it
CA2983282A1 (en) 2015-05-12 2016-11-17 Genentech, Inc. Therapeutic and diagnostic methods for cancer
JP7144935B2 (en) 2015-05-29 2022-09-30 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods for cancer
JP2018521019A (en) 2015-06-08 2018-08-02 ジェネンテック, インコーポレイテッド Method of treating cancer using anti-OX40 antibody
CA2988420A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
KR20180018695A (en) 2015-06-16 2018-02-21 가부시키가이샤 프리즘 파마 Anticancer drug
JP6896650B2 (en) 2015-06-17 2021-06-30 ジェネンテック, インコーポレイテッド Treatment of Locally Advanced or Metastatic Breast Cancer Using PD-1 Axle Antagonists and Taxanes
DK3341376T3 (en) 2015-08-26 2021-03-29 Fundacion Del Sector Publico Estatal Centro Nac De Investigaciones Oncologicas Carlos Iii F S P Cnio CONDENSED TRICYCLIC COMPOUNDS AS PROTEINKINASE INHIBITORS
BR112018005862A2 (en) 2015-09-25 2018-10-16 Genentech, Inc. antibodies, isolated antibodies, polynucleotide, vector, host cell, antibody production method, immunoconjugate, composition, uses of antibodies, methods for treating or retarding the progression of a cancer and a disease and for increasing, enhancing or stimulating a response or immune function and kit
CN108473435A (en) 2015-10-05 2018-08-31 纽约市哥伦比亚大学理事会 The treatment of the removing and protein sickness of the activator of autophagy tide and phospholipase D and the protein masses including TAU
CN108697713B (en) 2015-12-16 2021-10-26 基因泰克公司 Methods for making tricyclic PI3K inhibitor compounds and methods of treating cancer therewith
CA3006529A1 (en) 2016-01-08 2017-07-13 F. Hoffmann-La Roche Ag Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies
MX2018010361A (en) 2016-02-29 2019-07-08 Genentech Inc Therapeutic and diagnostic methods for cancer.
EP3865511A1 (en) 2016-04-14 2021-08-18 F. Hoffmann-La Roche AG Anti-rspo3 antibodies and methods of use
CA3019921A1 (en) 2016-04-15 2017-10-19 Genentech, Inc. Methods for monitoring and treating cancer
WO2017180581A1 (en) 2016-04-15 2017-10-19 Genentech, Inc. Diagnostic and therapeutic methods for cancer
EP3443120A2 (en) 2016-04-15 2019-02-20 H. Hoffnabb-La Roche Ag Methods for monitoring and treating cancer
JP7160688B2 (en) 2016-05-24 2022-10-25 ジェネンテック, インコーポレイテッド Heterocyclic inhibitors of CBP/EP300 and their use in treating cancer
JP7014736B2 (en) 2016-05-24 2022-02-01 ジェネンテック, インコーポレイテッド Pyrazolopyridine derivatives for the treatment of cancer
JP2019527037A (en) 2016-06-08 2019-09-26 ジェネンテック, インコーポレイテッド Diagnosis and treatment methods for cancer
US11046776B2 (en) 2016-08-05 2021-06-29 Genentech, Inc. Multivalent and multiepitopic antibodies having agonistic activity and methods of use
WO2018029124A1 (en) 2016-08-08 2018-02-15 F. Hoffmann-La Roche Ag Therapeutic and diagnostic methods for cancer
KR20190072528A (en) 2016-10-06 2019-06-25 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
JP2019535250A (en) 2016-10-29 2019-12-12 ジェネンテック, インコーポレイテッド Anti-MIC antibody and method of use
CA3048217A1 (en) 2016-12-22 2018-06-28 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2018160841A1 (en) 2017-03-01 2018-09-07 Genentech, Inc. Diagnostic and therapeutic methods for cancer
WO2018189220A1 (en) 2017-04-13 2018-10-18 F. Hoffmann-La Roche Ag An interleukin-2 immunoconjugate, a cd40 agonist, and optionally a pd-1 axis binding antagonist for use in methods of treating cancer
JOP20190272A1 (en) 2017-05-22 2019-11-21 Amgen Inc Kras g12c inhibitors and methods of using the same
FR3066761B1 (en) * 2017-05-23 2020-10-30 Centre Nat Rech Scient NEW IONIC CHANNEL INHIBITORS
TWI823859B (en) 2017-07-21 2023-12-01 美商建南德克公司 Therapeutic and diagnostic methods for cancer
WO2019033043A2 (en) 2017-08-11 2019-02-14 Genentech, Inc. Anti-cd8 antibodies and uses thereof
IL293443A (en) 2017-09-08 2022-07-01 Amgen Inc Inhibitors of kras g12c and methods of using the same
JP2020532982A (en) 2017-09-08 2020-11-19 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト How to diagnose and treat cancer
WO2019084395A1 (en) 2017-10-27 2019-05-02 University Of Virginia Patent Foundation Compounds and methods for regulating, limiting, or inhibiting avil expression
TW201923089A (en) 2017-11-06 2019-06-16 美商建南德克公司 Diagnostic and therapeutic methods for cancer
JP7391027B2 (en) 2018-02-26 2023-12-04 ジェネンテック, インコーポレイテッド Medication for treatment with anti-TIGIT and anti-PD-L1 antagonist antibodies
JP7266043B2 (en) 2018-05-04 2023-04-27 アムジエン・インコーポレーテツド KRas G12C inhibitors and methods of using them
JP7361722B2 (en) 2018-05-04 2023-10-16 アムジエン・インコーポレーテツド KRAS G12C inhibitors and methods of using the same
JP7361720B2 (en) 2018-05-10 2023-10-16 アムジエン・インコーポレーテツド KRAS G12C inhibitors for the treatment of cancer
JP2021524744A (en) 2018-05-21 2021-09-16 ナノストリング テクノロジーズ,インコーポレイティド Molecular gene signature and how to use it
ES2938987T3 (en) 2018-06-01 2023-04-18 Amgen Inc KRAS G12c inhibitors and methods of use thereof
JP7357644B2 (en) 2018-06-11 2023-10-06 アムジエン・インコーポレーテツド KRAS G12C inhibitors for treating cancer
MA51848A (en) 2018-06-12 2021-04-21 Amgen Inc KRAS G12C INHIBITORS AND THEIR PROCEDURES FOR USE
TWI819011B (en) 2018-06-23 2023-10-21 美商建南德克公司 Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
BR112020026641A2 (en) * 2018-06-27 2021-03-30 Oscotec Inc. PYRIDOPYRIMIDINONE DERIVATIVES FOR USE AS AXL INHIBITORS
JP2021530502A (en) 2018-07-18 2021-11-11 ジェネンテック, インコーポレイテッド How to Treat Lung Cancer with PD-1 Axial Binding Antagonists, Antimetabolites, and Platinums
CN110862398B (en) * 2018-08-27 2021-04-06 北京赛特明强医药科技有限公司 Urea substituted aromatic ring-linked dioxane quinazoline or quinoline compound, composition and application thereof
JP2021535169A (en) 2018-09-03 2021-12-16 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Carboxamide and sulfonamide derivatives useful as TEAD modulators
AU2019342099A1 (en) 2018-09-19 2021-04-08 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
ES2955032T3 (en) 2018-09-21 2023-11-28 Hoffmann La Roche Diagnostic methods for triple negative breast cancer
TW202028199A (en) * 2018-09-28 2020-08-01 大陸商重慶複創醫藥研究有限公司 Naphthyridinone and pyridopyrimidinone compounds useful as kinases inhibitors
CA3115038A1 (en) 2018-10-04 2020-04-09 Inserm (Institut National De La Sante Et De La Recherche Medicale) Egfr inhibitors for treating keratodermas
CA3116324A1 (en) 2018-10-18 2020-04-23 Genentech, Inc. Diagnostic and therapeutic methods for sarcomatoid kidney cancer
JP2020090482A (en) 2018-11-16 2020-06-11 アムジエン・インコーポレーテツド Improved synthesis of key intermediate of kras g12c inhibitor compound
MA55136A (en) 2018-11-19 2022-02-23 Amgen Inc G12C KRAS INHIBITORS AND METHODS OF USE THEREOF
JP7377679B2 (en) 2018-11-19 2023-11-10 アムジエン・インコーポレーテツド Combination therapy comprising a KRASG12C inhibitor and one or more additional pharmaceutically active agents for the treatment of cancer
US20220041579A1 (en) 2018-12-19 2022-02-10 Array Biopharma Inc. Substituted quinoxaline compounds as inhibitors of fgfr tyrosine kinases
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
AU2019403488A1 (en) 2018-12-20 2021-06-24 Amgen Inc. KIF18A inhibitors
WO2020132649A1 (en) 2018-12-20 2020-06-25 Amgen Inc. Heteroaryl amides useful as kif18a inhibitors
EA202191730A1 (en) 2018-12-20 2021-08-24 Эмджен Инк. KIF18A INHIBITORS
MX2021007157A (en) 2018-12-20 2021-08-16 Amgen Inc Heteroaryl amides useful as kif18a inhibitors.
JP2022519649A (en) 2019-02-08 2022-03-24 ジェネンテック, インコーポレイテッド How to diagnose and treat cancer
AU2020227319A1 (en) 2019-02-27 2021-09-09 Epiaxis Therapeutics Pty Ltd Methods and agents for assessing T-cell function and predicting response to therapy
JP2022521773A (en) 2019-02-27 2022-04-12 ジェネンテック, インコーポレイテッド Dosing for treatment with anti-TIGIT antibody and anti-CD20 antibody or anti-CD38 antibody
EP3931195A1 (en) 2019-03-01 2022-01-05 Revolution Medicines, Inc. Bicyclic heteroaryl compounds and uses thereof
WO2020180770A1 (en) 2019-03-01 2020-09-10 Revolution Medicines, Inc. Bicyclic heterocyclyl compounds and uses thereof
WO2020223233A1 (en) 2019-04-30 2020-11-05 Genentech, Inc. Prognostic and therapeutic methods for colorectal cancer
CN114269376A (en) 2019-05-03 2022-04-01 豪夫迈·罗氏有限公司 Methods of treating cancer with anti-PD-L1 antibodies
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
CN114144414A (en) 2019-05-21 2022-03-04 美国安进公司 Solid state form
CN110283171A (en) * 2019-07-17 2019-09-27 鼎泰(南京)临床医学研究有限公司 Compound of the one kind containing Pyridopyrimidine -4- amine structure, pharmaceutical composition and its application
CN112300279A (en) 2019-07-26 2021-02-02 上海复宏汉霖生物技术股份有限公司 Methods and compositions directed to anti-CD 73 antibodies and variants
MX2022001181A (en) 2019-08-02 2022-02-22 Amgen Inc Kif18a inhibitors.
AU2020326627A1 (en) 2019-08-02 2022-03-17 Amgen Inc. KIF18A inhibitors
EP4007753A1 (en) 2019-08-02 2022-06-08 Amgen Inc. Kif18a inhibitors
AU2020325115A1 (en) 2019-08-02 2022-03-17 Amgen Inc. Pyridine derivatives as KIF18A inhibitors
CR20220095A (en) 2019-09-04 2022-06-06 Genentech Inc Cd8 binding agents and uses thereof
CR20220127A (en) 2019-09-27 2022-05-27 Genentech Inc Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2021081212A1 (en) 2019-10-24 2021-04-29 Amgen Inc. Pyridopyrimidine derivatives useful as kras g12c and kras g12d inhibitors in the treatment of cancer
US20230024096A1 (en) 2019-10-29 2023-01-26 Hoffmann-La Roche Inc. Bifunctional compounds for the treatment of cancer
CN115873020A (en) 2019-11-04 2023-03-31 锐新医药公司 RAS inhibitors
KR20220109408A (en) 2019-11-04 2022-08-04 레볼루션 메디슨즈, 인크. RAS inhibitors
EP4055028A1 (en) 2019-11-04 2022-09-14 Revolution Medicines, Inc. Ras inhibitors
JP2022553803A (en) 2019-11-06 2022-12-26 ジェネンテック, インコーポレイテッド Diagnostic and therapeutic methods for the treatment of blood cancers
CN114901662A (en) 2019-11-08 2022-08-12 锐新医药公司 Bicyclic heteroaryl compounds and uses thereof
MX2022005775A (en) 2019-11-13 2022-06-09 Genentech Inc Therapeutic compounds and methods of use.
JP2023501528A (en) 2019-11-14 2023-01-18 アムジエン・インコーポレーテツド Improved Synthesis of KRAS G12C Inhibitor Compounds
TW202132271A (en) 2019-11-14 2021-09-01 美商安進公司 Improved synthesis of kras g12c inhibitor compound
EP4065231A1 (en) 2019-11-27 2022-10-05 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
EP4072584A1 (en) 2019-12-13 2022-10-19 Genentech, Inc. Anti-ly6g6d antibodies and methods of use
CN110903286B (en) * 2019-12-16 2021-09-24 沈阳药科大学 4, 6-disubstituted pyridine [3,2-d ] pyrimidine compound and preparation and application thereof
TW202136276A (en) 2019-12-20 2021-10-01 美商艾瑞斯卡公司 Tricyclic pyridones and pyrimidones
BR112022010086A2 (en) 2020-01-07 2022-09-06 Revolution Medicines Inc SHP2 INHIBITOR DOSAGE AND CANCER TREATMENT METHODS
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
EP4096646A1 (en) 2020-01-27 2022-12-07 Genentech, Inc. Methods for treatment of cancer with an anti-tigit antagonist antibody
WO2021177980A1 (en) 2020-03-06 2021-09-10 Genentech, Inc. Combination therapy for cancer comprising pd-1 axis binding antagonist and il6 antagonist
JP2023520515A (en) 2020-04-03 2023-05-17 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods for cancer
EP4143345A1 (en) 2020-04-28 2023-03-08 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
WO2021257503A1 (en) 2020-06-16 2021-12-23 Genentech, Inc. Methods and compositions for treating triple-negative breast cancer
MX2022015881A (en) 2020-06-18 2023-01-24 Genentech Inc Treatment with anti-tigit antibodies and pd-1 axis binding antagonists.
WO2021257736A1 (en) 2020-06-18 2021-12-23 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
CN116568824A (en) 2020-08-03 2023-08-08 基因泰克公司 Method for diagnosing and treating lymphoma
WO2022036146A1 (en) 2020-08-12 2022-02-17 Genentech, Inc. Diagnostic and therapeutic methods for cancer
JP2023541236A (en) 2020-09-03 2023-09-29 レボリューション メディシンズ インコーポレイテッド Use of SOS1 inhibitors to treat malignant tumors with SHP2 mutations
TW202227460A (en) 2020-09-15 2022-07-16 美商銳新醫藥公司 Ras inhibitors
AU2021347232A1 (en) 2020-09-23 2023-04-27 Erasca, Inc. Tricyclic pyridones and pyrimidones
TWI836278B (en) 2020-10-05 2024-03-21 美商建南德克公司 Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2022133345A1 (en) 2020-12-18 2022-06-23 Erasca, Inc. Tricyclic pyridones and pyrimidones
CN117396472A (en) 2020-12-22 2024-01-12 上海齐鲁锐格医药研发有限公司 SOS1 inhibitors and uses thereof
AU2022221124A1 (en) 2021-02-12 2023-08-03 F. Hoffmann-La Roche Ag Bicyclic tetrahydroazepine derivatives for the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
EP4334325A1 (en) 2021-05-05 2024-03-13 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
AU2022270116A1 (en) 2021-05-05 2023-12-21 Revolution Medicines, Inc. Ras inhibitors
AU2022280025A1 (en) 2021-05-25 2023-12-07 Erasca, Inc. Sulfur-containing heteroaromatic tricyclic kras inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
WO2023018699A1 (en) 2021-08-10 2023-02-16 Erasca, Inc. Selective kras inhibitors
AR127308A1 (en) 2021-10-08 2024-01-10 Revolution Medicines Inc RAS INHIBITORS
TW202332429A (en) 2021-11-24 2023-08-16 美商建南德克公司 Therapeutic compounds and methods of use
US20230203062A1 (en) 2021-11-24 2023-06-29 Genentech, Inc. Therapeutic compounds and methods of use
TW202340214A (en) 2021-12-17 2023-10-16 美商健臻公司 Pyrazolopyrazine compounds as shp2 inhibitors
WO2023129513A2 (en) * 2021-12-31 2023-07-06 President And Fellows Of Harvard College Bicyclic heteroaromatic inhibitors of nicotinamide n-methyltransferase, and compositions and uses thereof
WO2023154124A1 (en) * 2022-02-09 2023-08-17 Enliven Therapeutics, Inc. Acylated heterocyclic quinazoline derivatives as inhibitors of erbb2
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024033458A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydroazepine derivatives
WO2024033388A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives
WO2024033457A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives
WO2024033389A1 (en) 2022-08-11 2024-02-15 F. Hoffmann-La Roche Ag Bicyclic tetrahydrothiazepine derivatives
WO2024081916A1 (en) 2022-10-14 2024-04-18 Black Diamond Therapeutics, Inc. Methods of treating cancers using isoquinoline or 6-aza-quinoline derivatives
WO2024085242A2 (en) 2022-10-21 2024-04-25 Kawasaki Institute Of Industrial Promotion Non-fouling or super stealth vesicle
WO2024091991A1 (en) 2022-10-25 2024-05-02 Genentech, Inc. Therapeutic and diagnostic methods for multiple myeloma

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2949466A (en) * 1958-03-04 1960-08-16 Parke Davis & Co Pyrimidine compounds and means of producing the same
US3812127A (en) * 1966-10-31 1974-05-21 Pfizer 4-(quinolin-4-yl)piperazine-1-carboxylic acid esters
GB1199768A (en) * 1966-10-31 1970-07-22 Pfizer & Co C Nitrogen Heterocycles and process for their preparation
US3517005A (en) * 1967-10-26 1970-06-23 Pfizer & Co C Certain 2- and 4-substituted quinazolines
US3702849A (en) * 1967-10-26 1972-11-14 Pfizer 4-(isoquinolin-1-yl) piperazine-1-carboxylic acid esters
CH530412A (en) * 1968-07-15 1972-11-15 Ciba Geigy Ag Process for the preparation of (1) benzothieno- (2,3-d) pyrimidines
US3755583A (en) * 1970-06-05 1973-08-28 Chas0!nhx
FR2137271B1 (en) * 1971-05-11 1973-05-11 Progil
US3971783A (en) * 1973-03-07 1976-07-27 Pfizer Inc. 4-Aminoquinazoline derivatives as cardiac stimulants
GB1417029A (en) * 1973-03-07 1975-12-10 Pfizer Ltd Quinazoline-derived amines
SE435380B (en) * 1976-06-15 1984-09-24 Pfizer PROCEDURE FOR THE PREPARATION OF CHINAZOLINE COMPOUNDS
US4466965A (en) * 1982-07-26 1984-08-21 American Hospital Supply Corporation Phthalazine compounds, compositions and use
WO1984001151A1 (en) * 1982-09-24 1984-03-29 Beecham Group Plc Amino-azabicycloalkyl derivatives as dopamine antagonists
AU589409B2 (en) * 1985-03-14 1989-10-12 Sankyo Company Limited Phenoxyalkylaminopyrimidine derivatives, their preparation and insecticidal and acaricidal compositions containing them
US5141941A (en) * 1988-11-21 1992-08-25 Ube Industries, Ltd. Aralkylamine derivatives, and fungicides containing the same
JP2578000B2 (en) 1989-03-27 1997-02-05 株式会社石綿商店 Rotating door
GB8912336D0 (en) * 1989-05-30 1989-07-12 Smithkline Beckman Intercredit Compounds
US5034393A (en) * 1989-07-27 1991-07-23 Dowelanco Fungicidal use of pyridopyrimidine, pteridine, pyrimidopyrimidine, pyrimidopyridazine, and pyrimido-1,2,4-triazine derivatives
JPH03173872A (en) * 1989-09-14 1991-07-29 Ube Ind Ltd Aminopyrimidine derivative, preparation thereof and insecticidal-antimicrobial agent
IT1237141B (en) 1989-11-28 1993-05-24 PROFESSIONAL ESPRESSO COFFEE MACHINE FOR BARS
GB9022644D0 (en) * 1990-10-18 1990-11-28 Ici Plc Heterocyclic compounds
CA2095213A1 (en) * 1990-11-06 1992-05-07 Jotham W. Coe Quinazoline derivatives for enhancing antitumor activity
JP2762430B2 (en) * 1991-01-18 1998-06-04 宇部興産株式会社 Preparation of aralkylaminopyrimidines
ATE121735T1 (en) * 1991-02-20 1995-05-15 Pfizer 2,4-DIAMINOQUINAZOLINE DERIVATIVES TO INCREASE ANTITUMOR EFFECT.
WO1992020642A1 (en) * 1991-05-10 1992-11-26 Rhone-Poulenc Rorer International (Holdings) Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit egf and/or pdgf receptor tyrosine kinase
NZ243082A (en) * 1991-06-28 1995-02-24 Ici Plc 4-anilino-quinazoline derivatives; pharmaceutical compositions, preparatory processes, and use thereof
US5227387A (en) 1991-09-03 1993-07-13 Dowelanco Quinoline nematicidal method
DE4131029A1 (en) * 1991-09-18 1993-07-29 Basf Ag SUBSTITUTED PYRIDO (2,3-D) PYRIMIDINE AS ANTIDOTS
DE4131924A1 (en) * 1991-09-25 1993-07-08 Hoechst Ag SUBSTITUTED 4-ALKOXYPYRIMIDINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A PEST CONTROL
PT100905A (en) * 1991-09-30 1994-02-28 Eisai Co Ltd BICYCLE HYGIENEOUS HETEROCYCLIC COMPOUNDS CONTAINING BENZENE, CYCLOHEXAN OR PYRIDINE AND PYRIMIDINE, PYRIDINE OR IMIDAZOLE SUBSTITUTES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2681861B1 (en) 1991-10-01 1995-06-09 Isochem Sa PROCESS FOR THE PREPARATION OF A POLYMORPHIC FORM OF TERFENADINE, WITH HIGH MELTING POINT, OF GREATER PURITY AND PRODUCT OBTAINED.
US5187168A (en) * 1991-10-24 1993-02-16 American Home Products Corporation Substituted quinazolines as angiotensin II antagonists
AU661533B2 (en) * 1992-01-20 1995-07-27 Astrazeneca Ab Quinazoline derivatives
JP3173872B2 (en) 1992-05-15 2001-06-04 オリンパス光学工業株式会社 Optical element molding die and optical element molding method using the molding die
JPH0641134A (en) * 1992-07-21 1994-02-15 Nippon Soda Co Ltd Pyrimidopteridine derivative and its production
GB9323290D0 (en) * 1992-12-10 1994-01-05 Zeneca Ltd Quinazoline derivatives
TW370529B (en) * 1992-12-17 1999-09-21 Pfizer Pyrazolopyrimidines
JPH06220059A (en) * 1993-01-28 1994-08-09 Tanabe Seiyaku Co Ltd Condensed pyrimidine derivative and its production
DE4308014A1 (en) * 1993-03-13 1994-09-15 Hoechst Schering Agrevo Gmbh Condensed nitrogen heterocycles, processes for their preparation and their use as pesticides and fungicides
TW414798B (en) * 1994-09-07 2000-12-11 Thomae Gmbh Dr K Pyrimido (5,4-d) pyrimidines, medicaments comprising these compounds, their use and processes for their preparation
ATE201873T1 (en) * 1995-11-14 2001-06-15 Pharmacia & Upjohn Spa ARYL AND HETEROARYL PURINE AND PYRIDOPYRIMIDINE DERIVATIVES

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9512125B2 (en) 2004-11-19 2016-12-06 The Regents Of The University Of California Substituted pyrazolo[3.4-D] pyrimidines as anti-inflammatory agents
US20100143341A1 (en) * 2005-06-22 2010-06-10 Develogen Aktiengesellschaft Thienopyrimidines for pharmaceutical compositions
US8642604B2 (en) 2006-04-04 2014-02-04 The Regents Of The University Of California Substituted pyrazolo[3,2-d]pyrimidines as anti-cancer agents
US9493467B2 (en) 2006-04-04 2016-11-15 The Regents Of The University Of California PI3 kinase antagonists
US20100056548A1 (en) * 2006-04-07 2010-03-04 Develogen Aktiengesellschaft Thienopyrimidines having mnk1/mnk2 inhibiting activity for pharmaceutical compositions
US8633201B2 (en) 2006-04-07 2014-01-21 Boehringer Ingelheim International Gmbh Thienopyrimidines having Mnk1/Mnk2 inhibiting activity for pharmaceutical compositions
WO2007115822A1 (en) * 2006-04-07 2007-10-18 Develogen Aktiengesellschaft Thienopyrimidines having mnkl /mnk2 inhibiting activity for pharmaceutical compositions
US8697713B2 (en) 2006-07-10 2014-04-15 Boehringer Ingelheim International Gmbh Pyrrolopyrimidines for pharmaceutical compositions
US20100105708A1 (en) * 2006-07-10 2010-04-29 Develogen Aktiengesellschaft Pyrrolopyrimidines for pharmaceutical compositions
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US20110046165A1 (en) * 2008-01-04 2011-02-24 Pingda Ren Certain chemical entitles, compositions and methods
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US9655892B2 (en) 2008-01-04 2017-05-23 Intellikine Llc Certain chemical entities, compositions and methods
US9822131B2 (en) 2008-01-04 2017-11-21 Intellikine Llc Certain chemical entities, compositions and methods
US11433065B2 (en) 2008-01-04 2022-09-06 Intellikine Llc Certain chemical entities, compositions and methods
US9216982B2 (en) 2008-01-04 2015-12-22 Intellikine Llc Certain chemical entities, compositions and methods
US8703777B2 (en) 2008-01-04 2014-04-22 Intellikine Llc Certain chemical entities, compositions and methods
US8785456B2 (en) 2008-01-04 2014-07-22 Intellikine Llc Substituted isoquinolin-1(2H)-ones, and methods of use thereof
US20090312319A1 (en) * 2008-01-04 2009-12-17 Intellikine Certain chemical entities, compositions and methods
US9637492B2 (en) 2008-03-14 2017-05-02 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US20110124641A1 (en) * 2008-03-14 2011-05-26 Pingda Ren Benzothiazole kinase inhibitors and methods of use
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
US9828378B2 (en) 2008-07-08 2017-11-28 Intellikine Llc Kinase inhibitors and methods of use
US9629843B2 (en) 2008-07-08 2017-04-25 The Regents Of The University Of California MTOR modulators and uses thereof
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
US8486953B2 (en) 2008-08-26 2013-07-16 Boehringer Ingelheim International Gmbh Thienopyrimidines for pharmaceutical compositions
US9790228B2 (en) 2008-09-26 2017-10-17 Intellikine Llc Heterocyclic kinase inhibitors
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
US9296742B2 (en) 2008-09-26 2016-03-29 Intellikine Llc Heterocyclic kinase inhibitors
US8697709B2 (en) 2008-10-16 2014-04-15 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
US9315505B2 (en) 2009-05-07 2016-04-19 Intellikine Llc Heterocyclic compounds and uses thereof
US8785454B2 (en) 2009-05-07 2014-07-22 Intellikine Llc Heterocyclic compounds and uses thereof
US9206182B2 (en) 2009-07-15 2015-12-08 Intellikine Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US9522146B2 (en) 2009-07-15 2016-12-20 Intellikine Llc Substituted Isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US8569323B2 (en) 2009-07-15 2013-10-29 Intellikine, Llc Substituted isoquinolin-1(2H)-one compounds, compositions, and methods thereof
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US20110130706A1 (en) * 2009-11-19 2011-06-02 Follica, Inc. Sequential body surface treatment
US20110130711A1 (en) * 2009-11-19 2011-06-02 Follica, Inc. Hair growth treatment
US20110212102A1 (en) * 2010-02-26 2011-09-01 Boehringer Ingelheim International Gmbh Heterocycloalkyl-containing thienopyrimidines for pharmaceutical compositions
US20110212103A1 (en) * 2010-02-26 2011-09-01 Boehringer Ingelheim International Gmbh Thienopyrimidines containing a substituted alkyl group for pharmaceutical compositions
US20110217311A1 (en) * 2010-02-26 2011-09-08 Boehringer Ingelheim International Gmbh Cycloalkyl containing thienopyrimidines for pharmaceutical compositions
US8754079B2 (en) 2010-02-26 2014-06-17 Boehringer Ingelheim International Gmbh Cycloalkyl containing thienopyrimidines for pharmaceutical compositions
US8648068B2 (en) 2010-02-26 2014-02-11 Boehringer Ingelheim International Gmbh Heterocycloalkyl-containing thienopyrimidines for pharmaceutical compositions
US8853193B2 (en) 2010-02-26 2014-10-07 Boehringer Ingelheim International Gmbh Thienopyrimidines containing a substituted alkyl group for pharmaceutical compositions
US9181221B2 (en) 2010-05-21 2015-11-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9738644B2 (en) 2010-05-21 2017-08-22 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9388183B2 (en) 2010-11-10 2016-07-12 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8901133B2 (en) 2010-11-10 2014-12-02 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10550122B2 (en) 2011-01-10 2020-02-04 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one and methods of use thereof
US11312718B2 (en) 2011-01-10 2022-04-26 Infinity Pharmaceuticals, Inc. Formulations of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US9290497B2 (en) 2011-01-10 2016-03-22 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
USRE46621E1 (en) 2011-01-10 2017-12-05 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US9840505B2 (en) 2011-01-10 2017-12-12 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1 (2H)-one and methods of use thereof
US9295673B2 (en) 2011-02-23 2016-03-29 Intellikine Llc Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US9718815B2 (en) 2011-07-19 2017-08-01 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8969363B2 (en) 2011-07-19 2015-03-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9056877B2 (en) 2011-07-19 2015-06-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9605003B2 (en) 2011-07-19 2017-03-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9546180B2 (en) 2011-08-29 2017-01-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8785470B2 (en) 2011-08-29 2014-07-22 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9115141B2 (en) 2011-08-29 2015-08-25 Infinity Pharmaceuticals, Inc. Substituted isoquinolinones and methods of treatment thereof
US9321772B2 (en) 2011-09-02 2016-04-26 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US9895373B2 (en) 2011-09-02 2018-02-20 The Regents Of The University Of California Substituted pyrazolo[3,4-D]pyrimidines and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9527847B2 (en) 2012-06-25 2016-12-27 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
US11613544B2 (en) 2012-09-26 2023-03-28 The Regents Of The University Of California Substituted imidazo[1,5-a]pyrazines for modulation of IRE1
US10822340B2 (en) 2012-09-26 2020-11-03 The Regents Of The University Of California Substituted imidazolopyrazine compounds and methods of using same
US10131668B2 (en) 2012-09-26 2018-11-20 The Regents Of The University Of California Substituted imidazo[1,5-a]pYRAZINES for modulation of IRE1
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US11944631B2 (en) 2014-04-16 2024-04-02 Infinity Pharmaceuticals, Inc. Combination therapies
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies

Also Published As

Publication number Publication date
HRP950034A2 (en) 1997-10-31
MD1632F2 (en) 2001-03-31
US6521620B1 (en) 2003-02-18
US6265410B1 (en) 2001-07-24
FI114213B (en) 2004-09-15
US20030186987A1 (en) 2003-10-02
US6713484B2 (en) 2004-03-30
NZ281011A (en) 2002-02-01
PL315633A1 (en) 1996-11-25
SK89496A3 (en) 1997-10-08
RO117257B1 (en) 2001-12-28
PL179132B1 (en) 2000-07-31
AU686334B2 (en) 1998-02-05
CZ197096A3 (en) 1997-09-17
MD1632G2 (en) 2001-12-31
TJ381B (en) 2004-04-16
GEP20012376B (en) 2001-03-25
NO963094D0 (en) 1996-07-24
CN1493291A (en) 2004-05-05
BG100614A (en) 1997-03-31
MD960217A (en) 1998-04-30
US6084095A (en) 2000-07-04
FI20040648A (en) 2004-05-07
CN1140269C (en) 2004-03-03
FI20040649A (en) 2004-05-07
IL112249A (en) 2001-11-25
EP0742717A1 (en) 1996-11-20
FI962856A0 (en) 1996-07-15
WO1995019774A1 (en) 1995-07-27
JPH09508127A (en) 1997-08-19
CA2177372A1 (en) 1995-07-27
CN1139383A (en) 1997-01-01
BG63245B1 (en) 2001-07-31
HUT74589A (en) 1997-01-28
HU221741B1 (en) 2002-12-28
AU1731495A (en) 1995-08-08
HU9602017D0 (en) 1996-09-30
IL112249A0 (en) 1995-03-30
US6455534B2 (en) 2002-09-24
NO309892B1 (en) 2001-04-17
FI962856A (en) 1996-09-25
NO963094L (en) 1996-07-24

Similar Documents

Publication Publication Date Title
US6455534B2 (en) Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5654307A (en) Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6596726B1 (en) Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
EP1003745B1 (en) Naphthyridinones for inhibiting protein tyrosine kinase and cell cycle kinase mediated cellular proliferation
CZ286160B6 (en) 6-Arylpyrido[2,3-d]pyrimidines, their use and pharmaceutical preparations based thereon
SK283952B6 (en) Pyrido[2,3-d]pyrimidines for inhibiting protein tyrosine kinase mediated cellular proliferation
CA2177392C (en) Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family

Legal Events

Date Code Title Description
CC Certificate of correction
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20060924